Sélection de la langue

Search

Sommaire du brevet 2488346 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Brevet: (11) CA 2488346
(54) Titre français: PROCEDES ET COMPOSITIONS DESTINES A REPARER ET/OU A REGENERER UN MYOCARDE ENDOMMAGE
(54) Titre anglais: METHODS AND COMPOSITIONS FOR THE REPAIR AND/OR REGENERATION OF DAMAGED MYOCARDIUM
Statut: Périmé et au-delà du délai pour l’annulation
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • A61K 38/30 (2006.01)
  • A61K 38/18 (2006.01)
  • A61K 38/19 (2006.01)
  • A61P 21/00 (2006.01)
(72) Inventeurs :
  • ANVERSA, PIERO (Etats-Unis d'Amérique)
(73) Titulaires :
  • NEW YORK MEDICAL COLLEGE
(71) Demandeurs :
  • NEW YORK MEDICAL COLLEGE (Etats-Unis d'Amérique)
(74) Agent: SMART & BIGGAR LP
(74) Co-agent:
(45) Délivré: 2014-03-11
(86) Date de dépôt PCT: 2003-03-12
(87) Mise à la disponibilité du public: 2003-12-18
Requête d'examen: 2008-03-12
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/US2003/007731
(87) Numéro de publication internationale PCT: US2003007731
(85) Entrée nationale: 2004-12-03

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
10/162,796 (Etats-Unis d'Amérique) 2002-06-05

Abrégés

Abrégé français

Selon l'invention, des procédés, des compositions, et des nécessaires destinés à réparer des cellules myocardiques et/ou un myocarde endommagés, et notamment l'administration de cytokines, sont décrits et revendiqués.


Abrégé anglais


Methods, compositions, and kits for repairing damaged myocardium and/or
myocardial cells including the administration cytokines are disclosed and
claimed. Figure 2 shows a photograph of a tissue section from a myocardial
infarct (MI) induced mouse. Figure 2A-D show the area of MI INJECTED WITH Lin
negative/c-kit positive cells from bone marrow (arrow) at different
magnification, the remaining viable myocardium (VM), and the regenerating
myocardium (arrowheads). Figure 2E shows a photograph of a tissue section of
the area of MI injected with Lin negative/c-kit negative cells wherein only
healing is apparent.

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


93
I CLAIM:
1. Use of a cytokine capable of mobilizing c-kit POS adult cardiac stem
cells in the
manufacture of a medicament for repairing or regenerating damaged myocardium
in a heart,
wherein said medicament comprises multiple doses of the cytokine and wherein
the multiple
doses are formulated for administration by injection to provide a chemotactic
gradient of the
cytokine in the heart between a storage area of adult cardiac stem cells and
the damaged
myocardium, wherein the storage area of adult cardiac stem cells is one or
more of myocardial
apex, left atrium and right atrium, and wherein the cytokine is: stem cell
factor; insulin-like
growth factor-1 (IGF-1); hepatocyte growth factor (HGF); or granulocyte-
macrophage colony
stimulating factor (GMCSF).
2. Use of multiple doses of a cytokine capable of mobilizing c-kit POS
adult cardiac stem
cells and formulated for administration by injection to provide a chemotactic
gradient of the
cytokine in a heart between a storage area of adult cardiac stem cells and a
damaged
myocardium, for repairing or regenerating the damaged myocardium, wherein the
storage area
of adult cardiac stem cells is one or more of myocardial apex, left atrium and
right atrium,
wherein the cytokine is: stem cell factor; IGF-1; HGF; or GMCSF.
3. The use according to claim 1 or 2, wherein the multiple doses are for
administration to
different places in the heart between the storage area and a border zone of
the damaged
myocardium.
4. The use according to claim 1, 2 or 3, wherein amount of the cytokine
increases in a
direction towards the damaged myocardium.
5. The use according to any one of claims 1 to 4, wherein the chemotactic
gradient is
sufficient to cause adult cardiac stem cells resident in the heart to migrate
towards the damaged
myocardium.
6. The use according to any one of claims 1 to 5, wherein the multiple
doses comprise
different concentrations of the cytokine.
7. The use according to any one of claims 1 to 6, wherein the cytokine is
IGF-1.

94
8. The use according to any one of claims 1 to 6, wherein the cytokine is
HGF.
9. The use according to any one of claims 1 to 6, wherein the cytokine is
GMCSF.
10. The use according to any one of claims 1 to 5, wherein at least one
dose comprises a
plurality of cytokines capable of mobilizing stem cells.
11. The use according to any one of claims 1 to 5, wherein each dose
comprises a plurality
of cytokines capable of mobilizing stem cells.
12. The use according to claim 10 or 11, wherein the concentration of at
least one cytokine
in the plurality of cytokines is different in each dose.
13. The use according to claim 10, 11 or 12, wherein the concentration of
at least one
cytokine in the plurality of cytokines is the same in each dose.
14. The use according to any one of claims 10 to 13, wherein the plurality
comprises at
least one cytokine capable of inducing proliferation of the adult cardiac stem
cells.
15. The use according to any one of claims 10 to 14, wherein the plurality
of cytokines
comprise one or more of: stem cell factor; insulin-like growth factor-1 (IGF-
1); hepatocyte
growth factor (HGF); and granulocyte-macrophage colony stimulating factor.
16. The use according to claim 15, wherein at least one cytokine of the
plurality of
cytokines is IGF-1.
17. The use according to claim 15, wherein at least one cytokine of the
plurality of
cytokines is HGF.
18. The use according to claim 15, wherein the plurality of cytokines
comprises IGF-1 and
HGF.
19. The use according to claim 18, wherein the concentration of one of IGF-
1 and HGF is
varied between the multiple doses and the concentration of the other of IGF-1
and HGF is held
constant between the multiple doses.

95
20. The use according to claim 19, wherein the concentration of HGF is
varied between the
multiple doses and the concentration of IGF -1 is held constant between the
multiple doses.
21. The use according to any one of claims 7, 16 and 18 to 20, wherein the
multiple doses
each comprise IGF-1 at a concentration of up to about 500 ng/mL.
22. The use according to any one of claims 7, 16 and 18 to 20, wherein the
multiple doses
each comprise IGF-1 at a concentration of 150 to 250 ng/mL.
23. The use according to any one of claims 16 and 18 to 20, wherein the
multiple doses
each comprise IGF-1 at a concentration of about 200 ng/mL.
24. The use according to any one of claims 8 and 17 to 20, wherein the
multiple doses each
comprise HGF at a concentration of up to about 400 ng/mL.
25. The use according to any one of claims 8 and 17 to 20, wherein the
multiple doses each
comprise HGF at a concentration of 50 to 200 ng/mL.
26. The use according to any one of claims 1 to 25, wherein the multiple
doses are
formulated for administration via a catheter.
27. The use according to any one of claims 1 to 25, wherein the multiple
doses are
formulated for intramyocardial administration.
28. The use according to any one of claims 1 to 25, wherein the multiple
doses are
formulated for trans-epicardial administration.
29. The use according to any one of claims 1 to 25, wherein the multiple
doses are
formulated for transendocardial administration.
30. The use according to any one of claims 1 to 29, wherein said stem cells
are lineage
negative.

96
31. The use according to any one of claims 1 to 30, wherein the adult
cardiac stem cells are
capable of differentiating into myocytes, smooth muscle cells, or endothelial
cells in damaged
myocardium.
32. The use according to any one of claims 1 to 31, wherein the adult
cardiac stem cells
assemble into myocardial tissue.
33. The use of claim 32, wherein the tissue includes myocardial vessels.
34. The use according to any one of claims 1 to 33, wherein the adult
cardiac stem cells also
regenerate cardiac vessels.
35. A pharmaceutical composition comprising isolated adult cardiac stem
cells and a
pharmaceutically acceptable carrier, wherein said adult cardiac stem cells are
lineage negative,
c-kit positive, and express c-Met, IGF-1R receptor or both of said c-Met and
IGF-1R receptor.
36. The pharmaceutical composition of claim 35, wherein the cardiac stem
cells are
characterized by their ability to differentiate into myocytes, smooth muscle
cells, and
endothelial cells.
37. The pharmaceutical composition of claim 35 or 36, wherein the cardiac
stem cells are
human cardiac stem cells.
38. The pharmaceutical composition of claim 35, 36 or 37, wherein the
cardiac stem cells
are autologous.
39. The pharmaceutical composition of any one of claims 35 to 38, wherein
the
composition comprises 2 × 10 4 to 1 ×10 5 of said adult cardiac
stem cells.
40. The pharmaceutical composition of any one of claims 35 to 39, wherein
the
composition is formulated for intramyocardial or intraarterial injection.
41. The pharmaceutical composition of any one of claims 35 to 39, wherein
the
composition is formulated for catheter delivery to a heart.

97
42. The pharmaceutical composition of any one of claims 35 to 41, further
comprising one
or more cytokines.
43. The pharmaceutical composition of claim 42, wherein the one or more
cytokines are
hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1), or a
combination
thereof.
44. Use of isolated adult cardiac stem cells in manufacture of a medicament
for repairing or
regenerating a damaged myocardium, wherein the adult cardiac stem cells are
isolated from
myocardial tissue, and wherein said adult cardiac stem cells are c-kit
positive, lineage negative,
and express c-MET and/or IGF-1R receptor.
45. The use of claim 44, wherein the medicament further comprises one or
more cytokines.
46. The use of claim 45, wherein the one or more cytokines are hepatocyte
growth factor,
insulin-like growth factor-1, or a combination thereof.
47. Use of isolated adult cardiac stem cells for repairing or regenerating
a damaged
myocardium, wherein the adult cardiac stem cells are isolated from myocardial
tissue, and
wherein said adult cardiac stem cells are c-kit positive, lineage negative,
and express c-MET
and/or IGF-1R receptor.
48. The use according to any one of claims 44 to 47, wherein the isolated
adult cardiac stem
cells do not express markers of skeletal muscle cells, hematopoietic cells, or
neural cells when
exposed to differentiation medium in vitro.
49. The use according to any one of claims 44 to 48, wherein the isolated
adult cardiac stem
cells are capable of differentiating into myocytes, smooth muscle cells, and
endothelial cells.
50. The use according to any one of claims 44 to 49, wherein the isolated
adult cardiac stem
cells are autologous.
51. The use according to any one of claims 44 to 50, wherein the adult
cardiac stem cells
are formulated for administration by injection.

98
52. The use according to any one of claims 44 to 51, wherein the adult
cardiac stem cells
are formulated for administration via a catheter.
53. The use according to any one of claims 44 to 52, wherein the adult
cardiac stem cells
are formulated for intramyocardial administration.
54. The use according to any one of claims 44 to 52, wherein the adult
cardiac stem cells
are formulated for trans-epicardial administration.
55. The use according to any one of claims 44 to 52, wherein the adult
cardiac stem cells
are formulated for transendocardial administration.
56. The use according to any one of claims 44 to 52, wherein the adult
cardiac stem cells
are formulated for intraarterial administration.
57. The use of according to any one of claims 44 to 56, wherein the
isolated adult cardiac
stem cells are expanded in culture.
58. The use according to any one of claims 1 to 34 and 44 to 57, wherein
the damaged
myocardium is an infarction.
59. The pharmaceutical composition according to any one of claims 35 to 43,
for use in
treatment of a myocardial infarction.

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CA 02488346 2011-06-02
k
1
TITLE OF THE INVENTION
METHODS AND COMPOSITIONS FOR THE REPAIR AND/OR
REGENERATION OF DAMAGED MYOCARDIUM
10
20
There is
no admission that any of the various documents cited in this text are prior
art as to
the present invention. Any document having as an author or inventor person or
persons named as an inventor herein is a document that is not by another as to
the
inventive entity herein. Also, teachings of herein cited documents and
documents
cited in herein cited documents
can be employed in the practice and utilities of the present
invention.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
2
FIELD OF THE INVENTION
The present invention relates generally to the field of cardiology, and more
particularly relates to methods and cellular compositions for treatment of a
patient
suffering from a cardiovascular disease, including, but not limited to,
artherosclerosis, ischemia, hypertension, restenosis, angina pectoris,
rheumatic heart
disease, congenital cardiovascular defects and arterial inflammation and other
disease of the arteries, arterioles and capillaries.
Moreover, the present invention relates to any one or more of:
Methods and/or pharmaceutical composition comprising a therapeutically
effective amount of somatic stem cells alone or in combination with a cytokine
such
as a cytokine selected from the group consisting of stem cell factor (SCF),
granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage colony
stimulating factor (GM-CSF), stromal cell-derived factor-1, steel factor,
vascular
endothelial growth factor, macrophage colony stimulating factor, granulocyte-
macrophage stimulating factor or Interleukin-3 or any cytokine capable of the
stimulating and/or mobilizing stem cells. Cytokines may be administered alone
or
in combination of with any other cytokine capable of: the stimulation and/or
mobilization of stem cells; the maintenance of early and late hematopoiesis
(see
below); the activation of monocytes (see below), macrophage/monocyte
proliferation; differentiation, motility and survival (see below) and a
pharmaceutically acceptable carrier, diluent or excipient (including
combinations
thereof). The stem cells are advantageously adult stem cells, such as
hematopoietic
or cardiac stem cells or a combination thereof or a combination of cardiac
stem cells
and any other type of stem cells.
The implanting, depositing, administering or causing of implanting or
depositing or administering of stem cells, such as adult stem cells, for
instance
hematopoietic or cardiac stem cells or a combination thereof or any
combination of
cardiac stem cells (e.g., adult cardiac stem cells) and stem cells of another
type of
(e.g., adult stem cells of another type), alone or with a cytokine such as a
cytokine
selected from the group consisting of stem cell factor (SCF), granulocyte-
colony
stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor
(GM-CSF), stromal cell-derived factor-1, steel factor, vascular endothelial
growth

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
3
factor, macrophage colony stimulating factor, granulocyte-macrophage
stimulating
factor or Interleukin-3 or any cytokine capable of the stimulating and/or
mobilizing
stem cells (wherein "with a cytokine ..." can include sequential implanting,
depositing administering or causing of implanting or depositing or
administering of
the stem cells and the cytokine or the co-implanting co-depositing or co-
administering or causing of co-implanting or co-depositing or co-administering
or
the simultaneous implanting, depositing administering or causing of implanting
or
depositing or administering of the stem cells and the cytokine), in
circulatory tissue
or muscle tissue or circulatory muscle tissue, e.g., cardiac tissue, such as
the heart or
blood vessels ¨ e.g., veins, arteries, that go to or come from the heart such
as veins
and arteries directly connected or attached or flowing into the heart, for
instance the
aorta. This implanting, depositing, or administering or causing of implanting,
depositing or administering can be in conjunction with grafts. Such
implanting,
depositing or administering or causing of implanting, depositing or
administering is
advantageously employed in the treatment or therapy or prevention of cardiac
conditions, such as to treat areas of weakness or scarring in the heart or
prevent the
occurrence or further occurrence of such areas or to treat conditions which
cause or
irritate such areas, for instance myocardial infarction or ischemia or other
e.g.,
genetic, conditions that impart weakness or scarring to the heart (see also
cardiac
conditions mentioned infra).
The use of such stem cells alone or in combination with said cytokine(s), in
the formulation of medicaments for such treatment, therapy or prevention.
Medicaments for use in such treatment, therapy or prevention comprising the
stem cells and optionally the cytokine(s).
Kits comprising the stem cells and optionally the cytokine(s) for
formulations for use in such treatment, therapy or prevention.
Compositions comprising such stem cells and optionally at least one
cytokine and kits for preparing such compositions.
Methods of making the kits and compositions described herein.
Methods of implanting or depositing stem cells or causing the implanting or
depositing of stem cells.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
4
Methods and/or pharmaceutical compositions comprising a therapeutically
effective amount of one or more cytokines for causing the migration and/or
proliferation of cardiac stem cells or cardiac primative cells into
circulatory tissue or
muscle tissue or circulatory muscle tissue, e.g., cardiac tissue, such as the
heart or
blood vessels ¨ e.g., veins, arteries, that go to or come from the heart such
as veins
and arteries directly connected or attached or flowing into the heart, for
instance the
aorta. This migration and/or proliferation is advantageously employed in the
treatment or therapy or prevention of cardiac conditions, such as to treat
areas of
weakness or scarring in the heart or prevent the occurrence or further
occurrence of
such areas or to treat conditions which cause or irritate such areas, for
instance
myocardial infarction or ischemia or other e.g., genetic, conditions that
impart
weakness or scarring to the heart (see also cardiac conditions mentioned
infra).
Medicaments for use in such treatment, therapy or prevention comprising the
two or more cytokines.
Kits comprising the cytokines for formulations for use in such treatment,
therapy or prevention.
Compositions comprising the cytokines and kits for preparing such
compositions.
Methods of making the kits and compositions described herein.
Methods and/or pharmaceutical compositions comprising a therapeutically
effective amount of one or more cytokines for causing the migration and/or
proliferation of cardiac stem cells or cardiac primative cells into
circulatory tissue or
muscle tissue or circulatory muscle tissue, e.g., cardiac tissue, such_as the
heart or
blood vessels ¨ e.g., veins, arteries, that go to or come from the heart such
as veins
and arteries directly connected or attached or flowing into the heart, for
instance the
aorta in combination with a therapeutically effective amount of a
pharmaceutical
agent useful in treating hypertension, myocardial infarction, ischemia,
angina, or
other coronary or vascular ailments, such as ATi receptor blockers such as
losartan,
streptokinase, ReoPro (abciximab), enalapril maleate, Rapilysin (reteplase),
Dilatrend (carvedilol), Activase (alteplase), and other drugs for similar uses
which
would be known by one skilled in the art.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
Methods of treating a patient suffering from hypertension, myocardial
infarction, ischemia, angina or other coronary or vascular ailments, utilizing
the
above pharmaceutical compositions.
Kits comprising one or more cytokines in combination with a pharmaceutical
5 agent useful in treating hypertension, myocardial infarction, ischemia,
angina, or
other coronary or vascular ailments.
Methods of making and using the above kits and compositions.
BACKGROUND OF THE INVENTION
Cardiovascular disease is a major health risk throughout the industrialized
world. Atherosclerosis, the most prevalent of cardiovascular diseases, is the
principal cause of heart attack, stroke, and gangrene of the extremities, and
thereby
the principal cause of death in the United States. Atherosclerosis is a
complex
disease involving many cell types and molecular factors (for a detailed
review, see
Ross, 1993, Nature 362: 801-809).
Ischemia is a condition characterized by a lack of oxygen supply in tissues of
organs due to inadequate perfusion. Such inadequate perfusion can have number
of
natural causes, including atherosclerotic or restenotic lesions, anemia, or
stroke, to
name a few. Many medical interventions, such as the interruption of the flow
of
blood during bypass surgery, for example, also lead to ischemia. In addition
to
sometimes being caused by diseased cardiovascular tissue, ischemia may
sometimes
affect cardiovascular tissue, such as in ischemic heart disease. Ischemia may
occur
in any organ, however, that is suffering a lack of oxygen supply.
The most common cause of ischemia in the heart is myocardial infarction
(MI), commonly known as a heart attack, is one of the most well-known types of
cardiovascular disease. 1998 estimates show 7.3 million people in the United
States
suffer from MI, with over one million experiencing an MI in a given year
(American
Heart Association, 2000). Of these individuals, 25% of men, and 38% of females
will die within a year of their first recognized MI (American Heart
Association,
2000). MI is caused by a sudden and sustained lack of blood flow to an area of
the
heart, commonly caused by narrowing of a coronary artery. Without adequate
blood
supply, the tissue becomes ischemic, leading to the death of myocytes and
vascular

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
6
structures. This area of necrotic tissue is referred to as the infarct site,
and will
eventually become scar tissue. Survival is dependent on the size of this
infarct site,
with the probability of recovery decreasing with increasing infarct size. For
example, in humans, an infarct of 46% or more of the left ventricle triggers
irreversible cardiogenic shock and death (99).
Current treatments for MI focus on reperfusion therapy, which attempts to
start the flow of blood to the affected area to prevent the further loss of
tissue. The
main choices for reperfusion therapy include the use of anti-thrombolytic
agents, or
performing balloon angioplasty, or a coronary artery bypass graft. Anti-
thrombolytic agents solubilize blood clots that may be blocking the artery,
while
balloon angioplasty threads a catheter into the artery to the site of the
occlusion,
where the tip of the catheter is inflated, pushing open the artery. Still more
invasive
procedures include the bypass, where surgeons remove a section of a vein from
the
patient, and use it to create a new artery in the heart, which bypasses the
blockage,
and continues the supply of blood to the affected area. In 1998, there were an
estimated 553,000 coronary artery bypass graft surgeries and 539,000
percutaneous
transluminal coronary angioplastys. These procedures average $27,091 and
$8,982
per patient, respectively (American Heart Association, 2000).
These treatments may succeed in reestablishing the blood supply, however
tissue damage that occurred before the reperfusion treatment began has been
thought
to be irreversible. For this reason, eligible MI patients are started on
reperfusion
therapy as soon as possible to limit the area of the infarct.
As such, most studies on MI have also focused on reducing infarct size.
There have been a few attempts to regenerate the necrotic tissue by
transplanting
cardiomyocytes or skeletal myoblasts (Leor et al., 1996; Murray, et al., 1996;
Taylor, et al., 1998; Tomita et al., 1999; Menasche et al., 2000). While the
cells
may survive after transplantation, they fail to reconstitute healthy
myocardium and
coronary vessels that are both functionally and structurally sound.
All of the cells in the normal adult originate as precursor cells which reside
in various sections of the body. These cells, in turn, derive from very
immature
cells, called progenitors, which are assayed by their development into
contiguous
colonies of cells in 1-3 week cultures in semisolid media such as
methylcellulose or
agar. Progenitor cells themselves derive from a class of progenitor cells
called stem

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
7
cells. Stem cells have the capacity, upon division, for both self-renewal and
differentiation into progenitors. Thus, dividing stem cells generate both
additional
primitive stem cells and somewhat more differentiated progenitor cells. In
addition
to the well-known role of stem cells in the development of blood cells, stem
cells
also give rise to cells found in other tissues, including but not limited to
the liver,
brain, and heart.
Stem cells have the ability to divide indefinitely, and to specialize into
specific types of cells. Totipotent stem cells, which exist after an egg is
fertilized
and begins dividing, have total potential, and are able to become any type of
cell.
Once the cells have reached the blastula stage, the potential of the cells has
lessened,
with the cells still able to develop into any cell within the body, however
they are
unable to develop into the support tissues needed for development of an
embryo.
The cells are considered pluripotent, as they may still develop into many
types of
cells. During development, these cells become more specialized, committing to
give
rise to cells with a specific function. These cells, considered multipotent,
are found
in human adults and referred to as adult stem cells. It is well known that
stem cells
are located in the bone marrow, and that there is a small amount of peripheral
blood
stem cells that circulate throughout the blood stream (National Institutes of
Health,
2000).
Due to the regenerative properties of stem cells, they have been considered
an untapped resource for potential engineering of tissues and organs. It would
be an
advance to provide uses of stem cells with respect to addressing cardiac
conditions.
Mention is made of:
U.S. Patent No. 6,117,675 which relates to the differentiation of retinal stem
cells into retinal cells in vivo or in vitro, which can be used as a therapy
to restore
vision.
U.S. Patent No. 6,001,934 involving the development of functional islets
from islets of Langerhans stem cells.
U.S. Patents Nos.5,906,934 and 6,174,333 pertaining to the use of
mesenchymal stem cells for cartilage repair, and the use of mesenchymal stem
cells
for regneration of ligaments; for instance, wherein the stem cells are
embedded in a
gel matrix, which is contracted and then implanted to replace the desired soft
tissue.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
8
U.S. Patent Nos. 6,099,832, and 6,110,459 involving grafts with cell
transplantation.
PCT Application Nos. PCT/US00/08353 (WO 00/57922) and
PCT/US99/17326 WO 00/06701) involving intramyocardial injection of autologous
bone marrow and mesenchymal stern cells which fails to teach or suggest
administering, implanting, depositing or the use of hematopoietic stem cells
as in the
present invention, especially as hematopoietic stein cells as in the present
invention
are advantageously isolated and/or purified adult hematopoietic stem cells.
Furthermore, at least certain of these patent documents fail to teach or
suggest the present invention for additional reasons. The source of the stem
cells of
interest is limited to the known precursors of the type of tissue for which
regeneration is required. Obtaining and purifying these specific cells can be
extremely difficult, as there are often very few stem cells in a given tissue.
In
contrast, a benefit of the present invention results from the ability of
various lineages
of stem cells to home to the myocardium damage and differentiate into the
appropriate cell types- an approach that does not require that the stem cells
are
recovered directly from myocardium, and, a variety of types of stem cells may
be
used without compromising the functionality of the regenerated tissue. And,
other
of these patent documents utilize stem cells as the source of various chemical
compositions, without utilizing their proliferative capabilities, and thereby
fail to
teach or suggest the invention.
Only recent literature has started to investigate the potentials for stem
cells to
aid in the repair of tissues other than that of known specialization. This
plasticity of
stem cells, the ability to cross the border of germ layers, is a concept only
in its
infancy (Kempermann et al, 2000, Temple, 2001). Kocher et al (2001) discusses
the
use of adult bone marrow to induce neovascularization after infarction as an
alternative therapy for left ventricle remodeling (reviewed in Rosenthal and
Tsao,
2001). Other studies have focused on coaxing specific types of stem cells to
differentiate into myocardial cells, i.e. liver stem cells as shown in Malour
et al
(2001). Still other work focuses on the possibilities of bone-marrow derived
stem
cells (Krause, et al., 2001).
One of the oldest uses of stem cells in medicine is for the treatment of
cancer. In these treatments, bone marrow is transplanted into a patient whose
own

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
9
marrow has been destroyed by radiation, allowing the stem cells in the
transplanted
bone marrow to produce new, healthy, white blood cells.
In these treatments, the stem cells are transplanted into their normal
environment, where they continue to function as normal. Until recently, it was
thought that any particular stem cell line was only capable of producing three
or four
types of cells, and as such, they were only utilized in treatments where the
stem cell
was required to become one of the types of cells for which their ability was
already
proven. Researchers are beginning to explore other options for treatments of
myriad
disorders, where the role of the stem cell is not well defined. Examples of
such
work will be presented in support of the present invention.
Organ transplantation has been widely used to replace diseased,
nonfunctional tissue. More recently, cellular transplantation to augment
deficiencies
in host tissue function has emerged as a potential therapeutic paradigm. One
example of this approach is the well publicized use of fetal tissue in
individuals with
Parkinsonism (reviewed in Tompson, 1992), where dopamine secretion from
transplanted cells alleviates the deficiency in patients. In other studies,
transplanted
myoblasts from uneffected siblings fused with endogenous myotubes in
Duchenne's
patients; importantly the grafted myotubes expressed wild-type dystrophin
(Gussoni
et al., 1992).
Despite their relevance in other areas, these earlier studies do not describe
any cellular transplantation technology that can be successfully applied to
the heart,
where the ability to replace damaged myocardium would have obvious clinical
relevance. Additionally, the use of intra-cardiac grafts to target the long-
term
expression of angiogenic factors and ionotropic peptides would be of
therapeutic
value for individuals with myocardial ischemia or congestive heart failure,
respectively.
In light of this background there is a need for the improvement of myocardial
regeneration technology in the heart. Desirably, such technology would not
only
result in tissue regeneration in the heart but also enable the delivery of
useful
compositions directly to the heart. The present invention addresses these
needs.
It is therefore believed that heretofore the administration, implanting,
depositing, causing to be deposited, implanted or administered of stem cells,
alone
or in combination with at least one cytokine, as well as the use of such stem
cells

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
alone or in combination with said cytokine(s), in the formulation of
medicaments for
treatment, therapy or prevention, as in this disclosure and as in the present
invention,
has not been taught, or suggested in the art and that herein methods,
compositions,
kits and uses are novel, nonobvious and inventive, i.e., that the present
invention has
5 not been taught or suggested in the art and that the present invention is
novel,
nonobvious and inventive.
OBJECT AND SUMMARY OF THE INVENTION
It has surprisingly been found that the implantation of somatic stem cells
into
the myocardium surrounding an infarct following a myocardial infarction,
migrate
10 into the damaged area, where they differentiate into myocytes,
endothelial cells and
smooth muscle cells and then proliferate and form structures including
myocardium,
coronary arteries, arterioles, and capillaries, restoring the structural and
functional
integrity of the infarct.
It has also surprisingly been found that following a myocardial infarction,
the
administration of a cytokine to the patient, stimulates the patient's own
resident
and/or circulating stem cells, causing them to enter the blood stream and home
to the
infarcted area. It has also been found that once the cells home to the
infarct, they
migrate into the damaged tissue, where they differentiate into myocytes,
endothelial
cells and smooth muscle cells and then proliferate and form structures
including
myocardium, coronary arteries, arterioles and capillaries, restoring
structural and
functional integrity to the infracted area.
Surprisingly, resident cardiac stem cells (CSCs) have recently been identified
in the human (82) and rat (83, 84) heart. These primitive cells tend to
accumulate in
the atria (82) although they are also present throughout the ventricular
myocardium
(82, 83, 84). CSCs express surface antigens commonly found in hematopoietic
and
skeletal muscle stem cells (85, 86). CSCs are clonogenic, self-renewing and
multipotent giving rise to all cardiac lineages (84). Because of the growth
properties
of CSCs, the injured heart has the potential to repair itself. However, this
possibility
had been limited by our lack of understanding of CSC colonization,
proliferation and
differentiation in new organized, functioning myocardium (61, 87). Identical
obstacles apply to any other source of stem cells in the organism (88).

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
11
The identification of c-Met on hernatopoietic and hepatic stem cells (89, 90,
91) and, most importantly, on satellite skeletal muscle cells (92) has
prompted the
determining of whether its ligand, hepatocyte growth factor (HGF), has a
biological
effect on CSCs. Assuming that HGF mobilize and promote the translocation of
CSCs from anatomical storage areas to the site of damage acutely after
infarction.
HGF positively influences cell migration (93) through the expression and
activation
of matrix metalloproteinase-2 (94, 95). This enzyme family destroys barriers
in the
extracellular matrix thereby facilitating CSC movement, homing and tissue
restoration.
Similarly, insulin-like growth factor-1 (IGF-1) is mitogenic, antiapoptotic
and is necessary for neural stem cell multiplication and differentiation (96,
97, 98).
In a comparable manner, IGF-1 impacts CSCs by increasing their number and
protecting their viability. IGF-1 overexpression is characterized by myocyte
proliferation in the adult mouse heart (65) and this cell growth may depend on
CSC
activation, differentiation and survival.
Consequently, the invention provides methods and/or compositions for
repairing and/or regenerating recently damaged myocardium and/or myocardial
cells
comprising the administration of an effective amount of one or more cytokines,
e.g.
HGF and IGF-1 for causing the migration and/or proliferation of cardiac stem
cells
or cardiac primative cells into circulatory tissue or muscle tidssue or
circulatory
muscle tissue. This migration and/or proliferation is advantageously employed
in
the treatment or therapy or prevention of cardiac conditions, such as to treat
areas of
weakness or scarring in the heart or prevent the occurrence or further
occurrence of
such areas or to treat conditions which cause or irritate such areas, for
instance
myocardial infarction or ischemia or other, e.g. genetic, conditions that
impart
weakness or scarring to the heart.
It is reasonable to suggest that the protocol used here is superior to the
procedure employed to replace the necrotic or scarred myocardium by
transplanting
cardiomyocytes (42, 79), skeletal myoblasts (55, 76) or the prospective
utilization of
embryonic cells (100, 101). Although these attempts have been successful in
the
survival of many of the grafted cells, they have failed to reconstitute
healthy

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
12
myocardium and coronary vessels integrated structurally and functionally with
the
spared portion of the ventricular wall. CSCs are programmed to regulate the
normal
cell turnover of the heart and, under stressful conditions, participate in the
recovery
of the injured ventricle structurally and mechanically (82, 102).
The invention also provides methods and/or compositions comprising a
therapeutically effective amount of one or more cytokines for causing the
migration
and/or proliferation of cardiac stem cells or cardiac primative cells into
circulatory
tissue or muscle tissue or circulatory muscle tissue. This migration and/or
proliferation is advantageously employed in the treatment or therapy or
prevention
of cardiac conditions, such as to treat areas of weakness or scarring in the
heart or
prevent the occurrence or further occurrence of such areas or to treat
conditions
which cause or irritate such areas, for instance myocardial infarction or
ischemia or
other, e.g. genetic, conditions that impart weakness or scarring to the heart.
The invention also provides medicaments for use in such treatment, therapy
or prevention.
The invention further provides kits comprising one or more cytokines for
formulation for use in such treatment, therapy or prevention.
The invention still further provides methods of making the kits and
compositions described herein.
The invention further provides compositions and/or kits comprising one or
more cytokines in combination with a therapeutic agent for treating cardiac or
vascular conditions for formulation for use in such treatment, therapy or
prevention.
The invention provides to methods and/or compositions for repairing and/or
regenerating recently damaged myocardium and/or myocardial cells comprising
the
administration of somatic stem cells, e.g., adult stem cells or cardiac stem
cells or
hematopoietic stem cells or a combination thereof, such as adult cardiac or
adult
hematopoietic stem cells or a combination thereof or a combination of cardiac
stem
cells and a stem cell of another type, such as a combination of adult cardiac
stem
cells and adult stem cells of another type.
The invention further provides a method and/or compositions for repairing
and/or regenerating recently damaged myocardium and/or myocardial cells
comprising the administration of at least one cytokine.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
13
The invention further provides methods and/or compositions for repairing
and/or regenerating recently damaged myocardium and/or myocardial cells
comprising the administration of at least one cytokine in combination with a
pharmaceutical agent useful in the treatment of cardiac or vascular
conditions.
The invention still further relates to a method and/or compositions for
repairing and/or regenerating recently damaged myocardium comprising the
administration of somatic stem cells, e.g., adult stem cells or cardiac stem
cells or
hematopoietic stem cells or a combination thereof, such as adult cardiac or
adult
hematopoietic stem cells or a combination thereof or a combination of cardiac
stem
cells and a stem cell of another type, such as a combination of adult cardiac
stem
cells and adult stem cells of another type and a cytokine.
The invention yet further provides a method for preparing any of the
aforementioned or herein disclosed compositions comprising admixing the
pharmaceutically acceptable carrier and the somatic stem cells and/or
cytokines.
The invention also provides to a kit comprising a phaimaceutical
composition for use in repairing and/or regenerating recently damaged
myocardium
and/or myocardial cells.
The invention provides methods involving implanting, depositing,
administering or causing the implanting or depositing or administering of stem
cells,
such as adult stem cells, for instance hematopoietic or cardiac stem cells or
a
combination thereof or any combination of cardiac stein cells (e.g., adult
cardiac
stem cells) and stem cells of another type of (e.g., adult stem cells of
another type),
alone or with a cytokine such as a cytokine selected from the group consisting
of
stem cell factor (SCF), granulocyte-colony stimulating factor (G-CSF),
granulocyte-
macrophage colony stimulating factor (GM-CSF), stromal cell-derived factor-1,
steel factor, vascular endothelial growth factor, macrophage colony
stimulating
factor, granulocyte-macrophage stimulating factor or interleukin-3 or any
cytokine
capable of the stimulating and/or mobilizing stem cells (wherein "with a
cytokine
..." can include sequential implanting, depositing administering or causing of
implanting or depositing or administering of the stem cells and the cytokine
or the
co-implanting co-depositing or co-administering or causing of co-implanting or
co-
depositing or co-administering or the simultaneous implanting, depositing
administering or causing of implanting or depositing or administering of the
stem

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
14
cells and the cytokine), in circulatory tissue or muscle tissue or circulatory
muscle
tissue, e.g., cardiac tissue, such as the heart or blood vessels ¨ e.g.,
veins, arteries,
that go to or come from the heart such as veins and arteries directly
connected or
attached or flowing into the heart, for instance the aorta. This implanting,
depositing, or administering or causing of implanting, depositing or
administering
can be in conjunction with grafts.
Such implanting, depositing or administering or causing of implanting,
depositing or administering is advantageously employed in the treatment or
therapy
or prevention of cardiac conditions, such as to treat areas of weakness or
scarring in
the heart or prevent the occurrence or further occurrence of such areas or to
treat
conditions which cause or irritate such areas, for instance myocardial
infarction or
ischemia or other e.g., genetic, conditions that impart weakness or scarring
to the
heart (see also cardiac conditions mentioned supra).
The invention additionally provides the use of such stem cells alone or in
combination with said cytokine(s), in the formulation of medicaments for such
treatment, therapy or prevention.
And thus, the invention also provides medicaments for use in such treatment,
therapy or prevention comprising the stem cells and optionally the
cytokine(s).
Likewise the invention provides kits comprising the stem cells and optionally
the cytokine(s) for formulations for use in such treatment, therapy or
prevention.
The stem cells and the cytokine(s) can be in separate containers in a package
or in
one container in a package; and, the kit can optionally include a device for
administration (e.g., syringe) and/or instructions for administration and/or
admixture.
The invention also provides compositions comprising such stem cells and
optionally the cytokine(s) and kits for preparing such compositions (e.g.,
kits
comprising the stem cells and optionally the cytokine(s); stem cells and the
cytokine(s) can be in separate containers in a package or in one container in
a
package; and, the kit can optionally include a device for administration
(e.g.,
syringe) and/or instructions for administration and/or admixture), as well as
methods
of making the aforementioned compositions.
The invention also provides a means of generating and/or regenerating
myocardium ex vivo, wherein somatic stem cells and heart tissue are cultured
in

CA 02488346 2013-05-29
vitro, optionally in the presence of a cytokine. The somatic stem cells
differentiate into myocytes,
smooth muscle cells and endothelial cells, and proliferate in vitro, forming
myocardial tissue and/or
cells. These tissues and cells may assemble into cardiac structures including
arteries, arterioles,
capillaries, and myocardium. The tissue and/or cells formed in vitro may then
be implanted into a
5 patient, e.g. via a graft, to restore structural and functional
integrity.
Various embodiments of this invention provide use of a cytokine capable of
mobilizing c-
kitP s adult cardiac stem cells in the manufacture of a medicament for
repairing or regenerating
damaged myocardium in a heart, wherein said medicament comprises multiple
doses of the
cytokine and wherein the multiple doses are formulated for administration by
injection to provide a
10 chemotactic gradient of the cytokine in the heart between a storage area
of adult cardiac stem cells
and the damaged myocardium, wherein the storage area of adult cardiac stem
cells is one or more
of myocardial apex, left atrium and right atrium, and wherein the cytokine is:
stem cell factor;
insulin-like growth factor-1 (IGF-1); hepatocyte growth factor (HGF); or
granulocyte-macrophage
colony stimulating factor (GMCSF).
15 Various embodiments of this invention provide use of multiple doses of a
cytokine capable
of mobilizing c-kirs adult cardiac stem cells and formulated for
administration by injection to
provide a chemotactic gradient of the cytokine in a heart between a storage
area of adult cardiac
stem cells and a damaged myocardium, for repairing or regenerating the damaged
myocardium,
wherein the storage area of adult cardiac stem cells is one or more of
myocardial apex, left atrium
and right atrium, wherein the cytokine is: stem cell factor; IGF-1; HGF; or
GMCSF.
Various embodiments of this invention provide use of isolated adult cardiac
stem cells in
manufacture of a medicament for repairing or regenerating a damaged
myocardium, wherein the
adult cardiac stem cells are isolated from myocardial tissue, and wherein said
adult cardiac stem
cells are c-kit positive, lineage negative, and express c-MET and/or IGF-1R
receptor.
Various embodiments of this invention provide use of isolated adult cardiac
stem cells for
repairing or regenerating a damaged myocardium, wherein the adult cardiac stem
cells are isolated
from myocardial tissue, and wherein said adult cardiac stem cells are c-kit
positive, lineage
negative, and express c-MET and/or IGF-1R receptor.
Various embodiments of this invention provide a pharmaceutical composition
comprising
isolated adult cardiac stem cells and a pharmaceutically acceptable carrier,
wherein said adult
cardiac stem cells are lineage negative, c-kit positive, and express c-Met,
IGF-1R receptor or both
of said c-Met and IGF-1R receptor.

CA 02488346 2011-06-02
15a
In this disclosure, "comprises," "comprising," "containing" and "having" and
the like can have the meaning ascribed to them in U.S. Patent law and can mean
"includes," "including," and the like; "consisting essentially of' or
"consists
essentially" likewise has the meaning ascribed in U.S. Patent law and the
tennis
open-ended, allowing for the presence of more than that which is recited so
long as
basic or novel characteristics of that which is recited is not changed by the
presence
of more than that which is recited, but excludes prior art embodiments.
These and other embodiments are disclosed or are obvious from and
encompassed by, the following Detailed Description.
BRIEF DESCRIPTION OF FIGURES
The following Detailed Description, given to describe the invention by way
of example, but not intended to limit the invention to specific embodiments
described, may be understood in conjunction with the accompanying Figures,
incorporated herein by reference, in which:
Figure 1 shows a log-log plot showing Lin- bone marrow cells from EGFP
transgenic mice sorted by FACS based on c-kit expression (The fraction of c-
ki? s
cells (upper gate) was 6.4%. c-kit'EG cells are shown in the lower gate. c-ki?
s
cells were 1-2 logs brighter than c-kitNEG cells)
Figure 2A shows a photograph of a tissue section from a MI induced mouse
(The photograph shows the area of myocardial infarct (MI) injected with Lin-c-
ki? s
cells from bone marrow (arrows), the remaining viable myocardium (VM), and the
regenerating myocardium (arrowheads). Magnification is 12X);
Figure 2B shows a photograph of the same tissue section of Figure 2A at a
higher magnification, centering on the area of the MI with magnification being
50X;

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
16
Figures 2C, D show photographs of a tissue section at low and high
magnifications of the area of MI, injected with Lin- c-kirs cells, with the
magnification of 2C being 25X, and the magnification of 2D being 50 X;
Figure 2E shows a photograph of a tissue section of the area of MI injected
with Lin- c-kitNEG cells wherein only healing is apparent and the
magnification is
50X (*Necrotic myocytes. Red= cardiac myosin; green= PI labeling of nuclei);
Figures 3A-C show photographs of a section of tissue from a MI induced
mouse, showing the area of MI injected with Lin- c-kirs cells (Visible is a
section
of regenerating myocardium from endocardium (EN) to epicardium (EP). All
photographs are labeled to show the presence of infarcted tissue in the
subendocardium (IT) and spared myocytes in the subendocardium. (SM). Figure 3A
is stained to show the presence of EGFP (green). Magnification is 250X. Figure
3B
is stained to show the presence of cardiac myosin (red). Magnification is
250X.
Figure 3C is stained to show the presence of both EGFP and myosin (red-green),
as
well as PI-stained nuclei (blue). Magnification is 250X);
Figure 4A shows of grafts depicting the effects of myocardial infarction on
left ventricular end-diastolic pressure (LVEDP), developed pressure (LVDP), LV
+
rate of pressure rise (dP/dt), and LV - rate of pressure decay (dP/dt) (From
left to
right, bars indicate: sham-operated mice (SO, n=11); mice non-injected with
Lin- c-
kirs cells ( MI, n=5 injected with Lin- c-kitNEG cells; n=6 non-injected);
mice
injected with Lin- c-ki? s cells (MI+BM, n=9). Error bars are the standard
deviation. *' t p<0.05 vs SO and MI);
Figure 4B shows a drawing of a proposed scheme for Lin- c-kirs cell
differentiation in cardiac muscle and functional implications;
Figures 5A-I show photographs of a tissue sections from a MI induced
mouse depicting regenerating myocardium in the area of the MI which has been
injected with Lin- c-ki? s cells (Figure 5A is stained to show the presence of
EGFP
(green). Magnification is 300X. Figure 5B is stained to show the presence of a-
smooth muscle actin in arterioles (red). Magnification is 300X. Figure 5C is
stained

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
17
to show the presence of both EGFP and a-smooth muscle actin (yellow-red), as
well
as PI-stained nuclei (blue). Magnification is 300X. Figures 5D-F and G-I
depict
the presence of MEF2 and Csx/Nkx2.5 in cardiac myosin positive cells. Figure
5D
shows PI-stained nuclei (blue). Magnification is 300X. Figure 5E is stained to
show MEF2 and Csx/Nkx2.5 labeling (green). Magnification is 300X. Figure 5F is
stained to show cardiac myosin (red), as well as MEF2 or Csx/Nkx2.5 with PI
(bright fluorescence in nuclei). Magnification is 300X. Figure 5G shows PI-
stained
nuclei (blue). Magnification is 300X. Figure 5H is stained to show MEF2 and
Csx/Nkx2.5 labeling (green). Magnification is 300X. Figure 51 is stained to
show
cardiac myosin (red), as well as MEF2 or Csx/Nkx2.5 with PI (bright
fluorescence
in nuclei). Magnification is 300X);
Figure 6 (Figures 6A-F) shows photographs of tissue sections from MI
induced mice, showing regenerating myocardium in the area of the MI injected
with
Lin c-kites cells (Figures 6A-C show tissue which has been incubated in the
presence of antibodies to BrdU. Figure 6A has been stained to show PI-labeled
nuclei (blue). Magnification is 900X. Figure 6B has been stained to show BrdU-
and Ki67-labeled nuclei (green). Magnification is 900X. Figure 6C has been
stained to show the presence of a-sarcomeric actin (red). Magnification is
900X.
Figures 6D-F shows tissue that has been incubated in the presence of
antibodies to
K167. Figure 6D has been stained to show PI-labeled nuclei (blue).
Magnification
is 500X. Figure 6E has been stained to show BrdU- and Ki67-labeled nuclei
(green). Magnification is 500X. Figure 6F has been stained to show the
presence of
a-smooth muscle actin (red). Magnification is 500X. Bright fluorescence:
combination of PI with BrdU (C) or Ki67 (F));
Figure 7 (Figures 7A-C) shows photographs of tissue sections from MI
induced mice, showing the area of MI injected with Lin- c-kirs cells (Depicted
are
the border zone, viable myocardium (VM) and the new band (NB) of myocardium
separated by an area of infarcted non-repairing tissue (arrows). Figure 7A is
stained
to show the presence of EGFP (green). Magnification is 280X. Figure 7B is
stained

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
18
to show the presence of cardiac myosin (red). Magnification is 280X. Figure 7C
is
stained to show the presence of both EGFP and myosin (red-green), as well as
PI-
stained nuclei (blue). Magnification is 280X);
Figure 8 (Figures 8A-F) shows photographs of tissue sections from MI
induced mice, showing regenerating myocardium in the area of MI injected with
Lin c-kit' s cells (Figure 8A is stained to show the presence of EGFP (green).
Magnification is 650X. Figure 8B is stained to show the presence of cardiac
myosin
(red). Magnification is 650X. Figure 8C is stained to show both the presence
of
EGFP and myosin (yellow), as well as PI-stained nuclei (blue). Magnification
is
650X. Figure 8D is stained to show the presence of EGFP (green). Magnification
is
650X. Figure 8E is stained to show the presence of a-smooth muscle actin in
arterioles (red). Magnification is 650X. Figure 8F is stained to show the
presence
of both EGFP and a-smooth muscle actin (yellow-red) as well as PI-stained
nuclei
(blue). Magnification is 650X);
Figure 9 (Figures 9A-C) shows photographs of tissue sections from MI
induced mice, showing the area of MI injected with Lin- c-kirs cells and
showing
regenerating myocardium (arrowheads). (Figure 9A is stained to show the
presence
of cardiac myosin (red) Magnification is 400X. Figure 9B is stained to show
the
presence of the Y chromosome (green). Magnification is 400X. Figure 9C is
stained to show both the presence of the Y chromosome (light blue) and PI-
labeled
nuclei (dark blue). Note the lack of Y chromosome in infarcted tissue (IT) in
subendocardium and spared myocytes (SM) in subepicardium. Magnification is
400X);
Figure 10 (Figures 10A-C) shows photographs of tissue sections from MI
induced mice, showing GATA-4 in cardiac myosin positive cells (Figure 10A
shows PI-stained nuclei (blue). Magnification is 650X. Figure 10B shows the
presence of GATA-4 labeling (green). Magnification is 650X. Figure 10C is
stained to show cardiac myosin (red) in combination with GATA-4 and PI (bright
fluorescence in nuclei). Magnification is 650X);

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
19
Figure 11 (Figure 11A-D) shows photograph of tissue sections from a MI
induced mouse (Figure 11A shows the border zone between the infarcted tissue
and
the surviving tissue. Magnification is 500X. Figure 11B shows regenerating
myocardium. Magnification is 800X. Figure 11C is stained to show the presence
of
connexin 43 (yellow-green), and the contacts between myocytes are shown by
arrows. Magnification is 800X. Figure 11D is stained to show both a-sarcomeric
actin (red) and PI-stained nuclei (blue). Magnification is 800X);
Figure 12 (Figures 12A-B) shows photographs of tissue sections from a MI
induced mouse showing the area of MI that was injected with Lin- c-kirs cells
and
now shows regenerating myocytes (Figure 12A is stained to show the presence of
cardiac myosin (red) and PI-labeled nuclei (yellow-green). Magnification is
1,000.
Figure 12B is the same as Figure 12A at a magnification of 700X);
Figures 13A-B show photographs of tissue sections from MI induced mice
(Figure 13A shows a large infarct (MI) in a cytokine-treated mouse with
forming
myocardium (arrowheads) (Magnification is 50X) at higher magnification (80X -
adjacent panel). Figure 13B shows a MI in a non-treated mouse. Healing
comprises
the entire infarct (arrowheads) (Magnification is 50X). Scarring is seen at
higher
magnification (80X - adjacent panel). Red=cardiac myosin; yellow-
green=propidium iodide (PI) labeling of nuclei; blue-magenta=collagen types I
and
III);
Figure 13C shows a graph showing the mortality and myocardial
regeneration in treated and untreated MI induced mice (Cytokine-treated
infarcted
mice, n=15; untreated infarcted mice, n=52. Log-rank test: p<0.0001);
Figure 14 shows a graph showing quantitative measurement of infarct size
(Total number of myocytes in the left ventricular free wall (LVFW) of sham-
operated (SO, n=9), infarcted non-treated (MI, n=9) and cytokine-treated (MI-
C,
n=11) mice at sacrifice, 27 days after infarction or sham operation. The
percentage
of myocytes lost equals infarct size. X SD, *p<0.05 vs SO);

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
Figures 15A-C show graphs comparing aspects of myocardial infarction,
cardiac anatomy and function (Figures 15 A-C depict LV dimensions at
sacrifice,
27 days after surgery; sham-operated (SO, n=9), non-treated infarcted (MI,
n=9)
and cytokine-treated infarcted (MI-C, n=10));
5 Figure 15D
shows EF by echocardiography; (SO, n=9; MI, n=9; and MI-C,
n=9);
Figures 15E-M show M-mode echocardiograms of SO (e-g), MI (h-j) and
MI-C (k-m) (Newly formed contracting myocardium (arrows));
Figure 15N shows a graph showing wall stress; SO (n=9), MI (n=8) and MI-
10 C (n=9) (Results are mean SD. *'**p<0.05 vs SO and MI, respectively);
Figures 16A-G show grafts depicting aspects of myocardial infarction,
cardiac anatomy and ventricular function (Figures 16A-D show echocardiographic
LVESD (a), LVEDD (b), PWST (c) and PWDT (d) in SO (n=9), MI (n=9) and MI-
C (n=9). Figures 16E-G show mural thickness (e), chamber diameter (f) and
15 longitudinal axis (g) measured anatomically at sacrifice in SO (n=9), MI
(n=9) and
MI-C (n=10). ***p<0.05 vs SO and MI, respectively;
Figures 16H-P show two dimensional (2D) images and M-mode tracings of
SO (h-j), MI (k-m) and MI-C (n-p);
Figure 17 (Figures 17A-D) shows graphs depicting aspects of ventricular
20 function (Figure 17A-D show LV hemodynamics in anesthetized mice at
sacrifice,
27 days after infarction or sham operation; SO (n=9), MI (n=9) and MI-C
(n=10).
For symbols and statistics, see also Figure 13);
Figure 18A-E shows graphs of aspects of myocardial regeneration (Figure
18A classifies the cells in the tissue as remaining viable (Re), lost (Lo) and
newly
formed (Fo) myocardium in LVFW at 27 days in MI and MI-C; SO, myocardium
without infarct. Figure 18B shows the amount of cellular hypertrophy in spared
myocardium. Figure 18C shows cell proliferation in the regenerating
myocardium.
Myocytes (M), EC and SMC labeled by BrdU and K167; n=11. "*p<0.05 vs M and

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
21
EC. Figures 18D-E depict the volume, number (n=11) and class distribution
(bucket
size, 100 Am3; n=4,400) of myocytes within the formed myocardium;
Figures 18F-H show photographs of tissue sections from MI induced mice
depicting arterioles with TER-119 labeled erythrocyte membrane (green
fluorescence); blue fluorescence=PI staining of nuclei; red fluorescence=a-
smooth
muscle actin in SMC (Figure 18F is magnified at 800X. Figures 18G-H are
magnified at 1,200X);
Figure 19 (Figures 19A-D) shows photographs of tissue sections from MI
induced mice that were incubated with antibodies to Ki67 (A,B) and BrdU (C,D)
(Figure 19A shows labeling of myocytes by cardiac myosin. Bright fluorescence
of nuclei reflects the combination of PI and K167. Magnification is 800X.
Figure
19B shows labeling of SMC by a-smooth muscle actin. Bright fluorescence of
nuclei reflects the combination of PI and Ki67. Magnification is 1,200X.
Figure 19C shows labeling of SMC by a-smooth muscle actin. Bright fluorescence
of nuclei reflects the combination of PI and BrdU. Magnification is 1,200X.
Figure
19D shows labeling of EC in the forming myocardium by factor VIII. Bright
fluorescence of nuclei reflects the combination of PI and BrdU. Magnification
is
1,600X;
Figure 20 (Figures 20A-F) shows photographs of tissue sections from MI
induced mice showing markers of differentiating cardiac cells (Figure 20A is
stained to show labeling of myocytes by nestin (yellow)). Red fluorescence
indicates cardiac myosin. Magnification is 1,200X. Figure 20 B is stained to
show
labeling of desmin (red). Magnification is 800X. Figure 20C is stained to show
labeling of connexin 43 (green). Red fluorescence indicates cardiac myosin.
Magnification is 1,400X. Figure 20D shows VE-cadherin and yellow-green
fluorescence reflects labeling of EC by flk-1 (arrows). Magnification is
1,800X.
Figure 20E shows red fluorescence indicating factor VIII in EC and and yellow-
green fluorescence reflects labeling of EC by flk-1 (arrows). Magnification is
1,200X. Figure 20F shows green fluorescence labeling of SMC cytoplasms by flk-
1

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
22
and endothelial lining labeled by flk-1. Red fluorescence indicates a-smooth
muscle actin. Blue fluorescence indicates PI labeling of nuclei. Magnification
is
800X; and
Figure 21A-C show tissue sections from MI induced mice (Figure 21A uses
bright fluorescence to depict the combination of PI labeling of nuclei with
Csx/Nkx2.5. Magnification is 1,400X. Figure 21B uses bright fluorescence to
depict the combination of PI labeling of nuclei with GATA-4. Magnification is
1,200X. Figure 21C uses bright fluorescence to depict the combination of PI
labeling of nuclei with MBF2. Magnification is 1,200X (Red fluorescence shows
cardiac myosin antibody staining and blue fluorescence depicts PI labeling of
nuclei.
The fraction of myocyte nuclei labeled by Csx/Nkx2.5, GATA-4 and MEF2 was
63 5% (nuclei sampled=2,790; n=11), 94 9% (nuclei sampled=2,810; n=11) and
85*14% (nuclei sampled= 3,090; n=11), respectively).
Figure 22A-L are confocal micrographs which show cardiac primitive cells
in normal and growth factor-treated and untreated infarcted hearts. Figure 22A-
F
shows sections of atrial myocardium from sham-operated mice. Figure 22A and B,
22C and D, and 22E and F are pairs of micrographs showing the same area of
atrial
myocardium with different stains. c-Met (22A, yellow) is expressed in c-kitP s
(22B, green) cells (22B, yellow-green). Similarly, IGF-1R (22C, yellow) is
detected
in MDR1P s (22D, green) cells (22D, yellow-green). Colocalization of c-Met
(22E,
red) and IGF-1R (22E, yellow) are found in MDR1P s (22F, green) cells (22F,
red-
yellow-green). Arrows point to c-Met and IGF-1R in c-kit"s and MDR1P s cells.
Myocyte cytoplasm is stained red-purple and contains cardiac myosin. 22G: The
yellow line separates the infarcted myocardium (MI) with apoptotic myocytes
(bright nuclei, PI and hairpin 1) from the border zone (BZ) with viable
myocytes
(blue nuclei, PI only) in a mouse treated with growth factors. Viable c-IdtP s
cells
(blue nuclei, PI; c-kit, green) are present in MI and BZ (arrows). Myocyte
cytoplasm is stained red and contains cardiac myosin. 2211: The yellow line
separates the MI with necrotic myocytes (bright nuclei, PI and hairpin 2) from
the

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
23
BZ with viable myocytes (blue nuclei, PI only) in a mouse treated with growth
factors. Viable MDR11) s cells (blue nuclei, PI; MDR1, green) are present in
MI and
BZ (arrows). Myocyte cytoplasm is stained red and contains cardiac myosin).
221
and 22J: Apoptotic myocytes (221 and 22J, bright nuclei, PI and hairpin 1) and
c-
kit" (22!, green ring) and MDR1P s (22J, green ring) cells undergo apoptosis
(221
and 22J, bright nuclei, PI and hairpin 1; arrows) in the infarcted region of
two
untreated mice. Viable cells have blue nuclei (PI only). A viable c-kit' s
cell is
present within the infarcted myocardium (221, green ring, blue nucleus, PI
only;
arrowhead). Myocyte cytoplasm is stained red and shows cardiac myosin. 22K and
22L: Cycling c-kitP s (22K, green ring; arrows) and MDR1P s (22L, green ring;
arrows) cells are present in the infarcted myocardium (yellow dots are
apoptotic
nuclei) of mice treated with growth factors. Bright fluorescence in c-kit' s
(22K)
and MDR11) s (22L) cells corresponds to Ki67 labeling of their nuclei. 22A-L,
bar-10 m. 22M and 22N are graphs depicting the distribution of viable and
dead
c-kite s (22M) and MDR1P s (22N) cells in the various regions of the heart in
sham-
operated (SO), infarcted-treated (Treated) and infarcted-untreated (Untreated)
mice
sacrificed 7-8 hours after surgery and 2-3 hours after the administration of
growth
factors (Treated) or saline (SO; Untreated). Abbreviations are as follows: A,
atria;
LV, left ventricle; R, viable myocardium remote from the infarct; B, viable
myocardium bordering the infarct; I, non-viable infarcted myocardium. Results
in
both 22M and 22N are presented as the mean SD. *'** Indicates P < 0.05 vs.
SO
and vs. Treated, respectively.
Figure 23A-B are graphs depicting the size of infarct and the evaluation of
left ventricle hemodynamics. Results are presented as the mean SD. *'**
signifies
a value of p<0.05 vs. sham-operated mice (SO) and untreated infarcted mice
(MI),
respectively. Abbreviations are as follows: MI-T, treated infarcted mice; LV,
left
ventricle and septum. 23A: To minimize the effects of cardiac hypertrophy in
the
surviving myocardium and healing of the necrotic region with time on infarct
size,

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
24
infarct dimension was measured by the loss of myocytes in the left ventricle
and
septum. This measurement is independent from reactive hypertrophy in the
viable
tissue and shrinkage of the necrotic myocardium with scar formation (87). 23B:
Evaluation of LV hemodynamics is presented by data from LV end-diastolic
pressure, LV developed pressure, LV +dP/dt and LV ¨dP/dt. 23C to 23H are
confocal micrographs which depict large infarcts of the left ventricle in an
untreated
mouse (23C and 23D) and in two treated mice (23E to 2311). The area defined by
a
gate in 23C, 23E and 23G (bars=1 mm) is illustrated at higher magnification in
23D,
23F and 23H (bars=0.1 mm). In 23C and 23D the lack of myocardial regeneration
is illustrated by the accumulation of collagen type I and collagen type III
(blue) in
the infarcted region of the wall (arrows). Nuclei of spared myocytes and
inflammatory cells are apparent (green, PI). A small layer of viable myocytes
is
present in the subepicardium (red, cardiac myosin). In 23E to 2311, myocyte
regeneration is illustrated by the red fluorescence of cardiac myosin
antibody. Small
foci of collagen type I and type III (blue, arrowheads) are detected in the
infarcted
region. Nuclei are yellow-green (PI). Abbreviations are as follows: IS,
interventricular septum; MI, myocardial infarct; RV, right ventricle.
Figure 24 shows echocardiography results from a single mouse heart before
coronary artery ligation and 15 days after ligation. Confocal microscopy shows
a
cross section of the same heart. 24A shows the baseline echocardiography
results
before coronary artery ligation. 24B and 24C show confocal microscopy at low
(24B, bar = 1 mm) and higher (24C, bar = 0.1 mm) magnification of a cross
section
of the heart assessed in 24A and 24D. Abbreviations used are as follows: RV,
right
ventricle; IS, interventricular septum; MI, myocardial infarct. 24D shows the
echocardiographic documentation of contractile function in the same heart 15
days
after infarction (arrowheads). 24E is a graph depicting the ejection fraction
with
results reported as the mean SD. *'** p<0.05 vs. sham-operated mice (SO) and
untreated infarcted mice (MI), respectively. MI-T refers to treated infarcted
mice.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
Figure 25A-F shows confocal micrographs detailing properties of
regenerating myocytes. These properties are quantified in the graphs of 25G-J.
25A and 25B depict enzymatically dissociated myocytes from the regenerating
portion (25A) and surviving myocardium (25B) of the infarcted ventricle of a
heart
5 treated with growth factors. 25A is stained to show small myocytes (red,
cardiac
myosin), bright nuclei (PI and BrdU), and blue nuclei (PI only). 25B shows
large,
hypertrophied myocytes (red, cardiac myosin), bright nuclei (PI and BrdU) and
blue
nuclei (PI only). In both 25A and 25B, the bar equals 50 IAM. Mechanical
properties of new (25C and 25D) and spared (25E and 25F) myocytes are shown
10 after infarction in mice treated with growth factors. R refers to the
relaxed state of
they myocytes, C is the contracted state. The effects of stimulation on cell
shortening (G), velocity of shortening (H), time to peak shortening (I) and
time to
50% re-lengthening (J) are depicted with results given for N (new small
myocytes)
and S (spared hypertrophied myocytes). Results are presented as the mean SD. *
15 indicates a value of P<0.05 vs S.
Figure 26 shows pairs of confocal micrographs showing various markers of
maturing myocytes (26A to 26N, bar=10 In 26A to 26F, BrdU labeling of
nuclei is shown in 26A, 26C and 26E as green coloration, and localization of
nestin
(26B, red), desmin (26D, red), cardiac myosin (26F, red) is shown in myocytes
of
20 tissue sections of regenerating myocardium. Nuclei are labeled by PI
only in 26B,
26D and 26F (blue), and by BrdU and PI together in 26B, 26D and 26F (bright).
26G to 26N show the identification of connexin 43 (26G, 2611, 26K and 26L,
yellow) and N-cadherin (261, 26J, 26M and 26N, yellow) in sections of
developing
myocardium (26G to 26J) and in isolated myocytes (26K to 26N). Myocytes are
25 stained by cardiac myosin (26H, 26J, 26L and 26N, red) and nuclei by
BrdU only
(26G, 261,26K and 26M, green), PI only (2611 and 26J, blue) and by BrdU and PI
together (2611, 26J, 26L and 26N, bright).

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
26
Figure 27 is a series of confocal micrographs showing newly formed
coronary vasculature. In 27A to 27D, arterioles are shown with TER-119-labeled
erythrocyte membrane (green), PI staining of nuclei (blue), and a-smooth
muscle
actin staining of smooth muscle cell (red). In all micrographs, the bar equals
10 Inn.
Figure 28: Identification and growth of cardiac Lin-c-kiPs cells obtained
with immunomagnetic beads (a) and FACS (b). a,b, c-kirs cells in NSCM scored
negative for cytoplasmic proteins of cardiac cell lineages; nuclei are stained
by PI
(blue) and c-kit (green) by c-kit antibody. c-f, In DM at Pl, cultured cells
showed by
purple fluorescence in their nuclei Nkx2.5 (c), MEF2 (d), GATA-4 (e) and GATA-
5
(f) labeling. g,h, Stem cells selected by NSCM and plated at low density (g)
develop
small individual colonies (h). Bar=10 m.
Figure 29: Self-renewal and multipotentiality of clonogenic cells. a, c-kirs
cells in a clone: nuclei¨blue, c-kit=green (arrowheads). b, Two of the 3 c-
kite's cells
(green, arrowheads) express Ki67 (purple, arrows) in nuclei (blue). c,d, Ki67
positive (c) metaphase chromosomes (red). d, metaphase chromosomes labeled by
Ki67 and PI (purple) in a c-ki? s cell (green). e-h, In the clone, the
cytoplasm (red)
of M (e), EC (f), SMC (g) and F (h) is stained by cardiac myosin, factor VIII,
a-
smooth muscle actin and vimentin, respectively. Nuclei=blue. Lin-c-kitP s
cells
(green, arrowheads) are present. Bar=10 m.
Figure 30: Clonogenic cells and spherical clones. a, Spherical clones
(arrowheads) in suspension in NSCM. b, Cluster of c-kit' s (green, arrowheads)
and
negative cells within the clone. Nuclei=blue. c, Spheroid with packed cell
nuclei
(blue) and large amount of nestin (red). d, Accumulation of non-degraded
nestin
(red) within the spheroid. Nuclei=blue. e, Spheroid plated in DM with cells
migrating out of the sphere. f-h, M (f), SMC (g) and EC (h) migrating out of
the
spheroid and differentiating have the cytoplasm (red) stained respectively by
cardiac
myosin, a-smooth muscle actin and factor VIII. Nuclei=blue. Bar=10ium.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
27
Figure 31: Myocardial repair. a-c, Generating myocardium (a,b,
arrowheads) in an infarcted treated rat (MI). New M=myosin (red); nuclei=
yellow-
green. Sites of injection (arrows). c, Myocardial scarring (blue) in an
infarcted
untreated rat. *Spared myocytes. d-1, M (f, myosin) and coronary vessels (i,
EC=factor VIII; 1, SMC=a-smooth muscle actin) are identified by BrdU (green)
positive nuclei (e,h,k). Blue nuclei¨PI (d,g,j). m-t, Myocytes at 20 days
(o,p,s,t) are
more differentiated than at 10 (m,n,q,r). m-p: connexin 43=yellow
(arrowheads), q-
t: N-cadherin=yellow (arrowheads); Myosin=red. Nuclei= blue; BrdU=green
(arrows). Bar=1 mm (a), 100 m (b,c), 1011111 (d-t).
Figure 32: Newly generated myocytes. a, enzymatically dissociated cells
from the repairing myocardial band. Cardiac myosin=red; Brdu=green;
nuclei=blue.
b-e, differentiation of new myocytes. Connexin 43=yellow (b,c); N-
cadherin=yellow (d,e). Cardiac myosin-- red; Brdu=green; nuclei=blue. Bar=10
m.
Figure 33: Mechanical properties of myocytes. a-d, new (N) and spared (S)
myocytes obtained, respectively, from the regenerating and remaining
myocardium
after infarction in treated rats; R=relaxed, C=contracted. e-h, effects of
stimulation
on cell shortening and velocity of shortening of N (e,g) and S (f,h) myocytes.
1-1,
Results are mean SD. *P<0.05 vs S.
Figure 34: Primitive Cells in the Rat Heart. Section of left ventricular
myocardium from a Fischer rat at 22 months of age. A, Nuclei are illustrated
by the
blue fluorescence of propidium iodide (PI). B, Green fluorescence documents c-
kit
positive cells. C, The combination of PI and c-kit is shown by green and blue
fluorescence. The myocyte cytoplasm is recognized by the red fluorescence of
ct-
sarcomeric actin antibody staining. Confocal microscopy; bar=10 pm.
Figure 35: FACS Analysis of c-ki? s Cells. Bivariate distribution of
cardiac cells obtained from the left ventricle of a female Fischer 344 rat
showing the
level of c-kit expression versus cellular DNA. The cells were suspended at a
concentration of 106 cells/ml of PBS. Cellular fluorescence was measured with
the

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
28
ELITE ESP flow cytometer/cell sorter (Coulter Inc.) using an argon ion laser
(emission at 488 nm) combined with a helium-cadmium laser, emitting UV light.
Arrow indicates a threshold representing minimal c-kit level. For FACS
analysis,
cells were incubated with r-phycoerythrin (R-PE)-conjugated rat monoclonal c-
kit
antibody (Pharmingen). R-PE isotype standard was used as a negative control.
Figure 36: Scheme for Collection of Cardiac c-kite s Cells (A) and
Culture of Cardiac c-ki? s Cells in NSCM (B). A, Undifferentiated cells
expressing c-kit surface receptors are exposed to c-kit antibody and
subsequently to
immunomagnetic beads coated by IgG antibody. c-ki?Gs cells are collected with
a
magnet and cultured in NSCM. B, Immunomagnetic beads are attached on the
surface of c-ki?Gs cells (arrowheads). The absence of c-kitNEG cells is
apparent.
Phase contrast microscopy; bar=101.tm.
Figure 37: c-kit Protein in Freshly Isolated Cells Collected with
Immunomagnetic Beads. c-kit protein is shown by the green fluorescence of c-
kit
antibody. Beads adherent to the cells are illustrated by red fluorescence.
Blue
fluorescence reflects PI labeling of nuclei. Thus, cells selected with beads
were
found to be c-ki?Gs. Confocal microscopy; bar=10 i.im.
Figure 38: Transcription Factors of Cardiomyocyte Differentiation.
After removal of the beads, or immediately after FACS separation, smears were
made and cells were stained for the detection of NIcx2.5, MEF2 and GATA-4.
Blue
fluorescence in panels A-C corresponds to PI labeling of nuclei. Purple
fluorescence
in nuclei reflects the expression of NIcx2.5 (A), MEF2 (B) and GATA-4 (C).
Confocal microscopy; bar=10
Figure 39: c-ki? s Cells and Transcription Factors of Skeletal Muscle
Differentiation. Panels A-C shows c-kiPs cells (green fluorescence, c-kit
antibody;
blue fluorescence, PI labeling). Panels D-F illustrate positive controls
(C2C12
myoblast cell line) for MyoD (D), myogenin (E), and Myf5 (F) by green
fluorescence within nuclei (red fluorescence, PI labeling). c-kit' s cells
were

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
29
negative for these skeletal muscle transcription factors. Confocal microscopy;
bar=10 [un.
Figure 40: Growth of c-kiPs Cells in Differentiating Medium (DM).
Monolayer of confluent cells obtained from plating c-kit positive cells.
Immunomagnetic beads were removed by gentle digestion of the cells with DNase
I.
This procedure degraded the short DNA linker between the bead and the anti-IgG
antibody. Phase contrast microscopy; bar=20
Figure 41: Cycling Cell Nuclei in DM. Ki67 (purple fluorescence) is
expressed in the majority of nuclei contained in the field. Blue fluorescence
reflects
PI labeling of nuclei. Confocal microscopy; bars=10
Figure 42: Growth Rate of c-kitP s-Derived Cells. Exponential growth
curves of cells at P2 and P4; -ID, time required by the cells to double in
number. Each
point corresponds to 5 or 6 independent determinations. Vertical bars, SD.
Figure 43: Identification and Growth of Cardiac Lin-c-kiPs Cells. In
DM at P3, the cytoplasm (green) of M (A), EC (B), SMC (C) and F (D) is stained
by
cardiac myosin, factor VIII, a-smooth muscle actin and vimentin (factor VIII
negative), respectively. Nuclei= red. Confocal microscopy; bars=10
Figure 44: Cytoplasmic Markers of Neural Cells. Panels A-C shows cells
in DM at P1 (red fluorescence, a-sarcomeric actin; blue fluorescence, PI
labeling).
Panels D-F illustrate positive controls for MAP lb (D, neuron2A cell line),
nerofilament 200 (E, neuron2A cell line), and GFAP (F, astrocyte type III,
clone C8-
D30) by green fluorescence in the cytoplasm (blue fluorescence, PI labeling).
c-
ki? s-derived cells were negative for these neural proteins. Confocal
microscopy;
bar=10
Figure 45: Cytoplasmic Markers of Fibroblasts. Panels A-C shows small
colonies of undifferentiated cells in NSCM (green fluorescence, c-kit; blue
fluorescence, PI labeling). Panels D-F illustrate positive controls (rat heart
fibroblasts) for fibronectin (D), procollagen type I (E), and vimentin (F) by
red

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
fluorescence in the cytoplasm (blue fluorescence, PI labeling). c-kiPs-derived
cells
were negative for these fibroblast proteins. Confocal microscopy; bar=10
Figure 46: FACS-Isolated c-ki? s Cells: Multipotentiality of Clonogenic
Cells. In a clone, the cytoplasm (red) of M (A), EC (B), SMC (C) and F (D) is
5 stained by cardiac myosin, factor VIII, a-smooth muscle actin and
vimentin,
respectively. Blue fluorescence, PI labeling of nuclei. Lin-c-kitP s cells
(green
fluorescence, arrowheads) are present. Confocal microscopy; bar=10 pm.
Figure 47: Cardiac Cell Lineages in Early Differentiation. A,B,
Expression of nestin alone (green fluorescence) in the cytoplasm of cells in
early
10 differentiation. C,D, Expression of nestin (green, C) and cardiac myosin
(red, D) in
developing myocytes (arrowheads). E,F, Expression of nestin (green, E) and
factor
VIII (red, F) in developing endothelial cells (arrowheads). G,H, Expression of
nestin
(green, G) and a-smooth muscle actin (red, H) in developing smooth muscle
cells
(arrowheads). Confocal microscopy; bars=10 m.
15 Figure 48: Infarct Size and Myocardial Repair. A, At 10 days, coronary
artery occlusion resulted in the loss of 49% and 53% of the number of myocytes
in
the left ventricle of untreated (MI) and treated (MI-T) rats, respectively. At
20 days,
coronary artery occlusion resulted in the loss of 55% and 70% of the number of
myocytes in the left ventricle of untreated (MI) and treated (MI-T) rats,
respectively.
20 SO, sham-operated animals. *P<0.05 vs SO. t/3<0.05 vs MI. B, Percentage
of newly
formed myocardium within the infarcted region of the wall at 10 and 20 days
(d)
after coronary artery occlusion in animals treated with cell implantation (MI-
T).
*P<0.05 vs 10d. C,D, The amount of new myocardium formed (F) at 10 and 20 days
by cell implantation was measured morphometrically (solid bar). The remaining
(R)
25 and lost (L) myocardium after infarction is depicted by hatched bar and
crosshatched
bar, respectively. The generated tissue (F) increased the remaining myocardium
(R+F) and decreased the lost myocardium (L-F) by the same amount. As a
consequence, cardiac repair reduced infarct size in both groups of rats
treated with

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
31
cell implantation. Results are mean SD. *P<0.05 vs MI. 1P<0.05 vs Lo and Fo in
MI-T.
Figure 49: Myocardial Repair. A,B, Bands of regenerating myocardium in
two infarcted treated hearts. Red fluorescence corresponds to cardiac myosin
antibody staining of newly formed myocytes. Yellow-green fluorescence reflects
PI
labeling of nuclei. Blue fluorescence (arrowheads) illustrates small foci of
collagen
accumulation within the infarcted region of the wall. Confocal microscopy;
bar=100
gm.
Figure 50: Neoformation of Capillaries. The differentiation of implanted
cells in capillary profiles was identified by BrdU labeling of endothelial
cells. A, PI
labeling of nuclei (blue); B, BrdU labeling of nuclei (green); C, Capillary
endothelium (red) and endothelial cell nuclei labeled by BrdU (blue and
green).
Confocal microscopy; bar=10 gm.
Figure 51: Volume Composition of Regenerating Myocardium. During
the interval from 10 to 20 days, the volume fraction of myocytes (M),
capillaries
(Cap) and arterioles (Art) increased 25%, 62% and 140%, respectively.
Conversely,
the volume percent of collagen type I (C-I) and collagen type III (C-ill)
decreased
73% and 71%, respectively. Results are mean SD. *P<0.05 vs 10 days.
Figure 52: Cell Proliferation in the Regenerating Myocardium. During
the interval from 10 to 20 days, the fraction of myocytes (M), endothelial
cells (EC)
and smooth muscle cells (SMC) labeled by Ki67 decreased 64%, 63% and 59%
respectively. Results are mean SD. *P<0.05 vs 10 days.
Figure 53: Identification of Regenerating Myocytes by BrdU Labeling.
A,D, Nuclei are illustrated by the blue fluorescence of PI. B,E, Green
fluorescence
documents BrdU labeling of nuclei. C,F, Myocyte cytoplasm is recognized by the
red fluorescence of a-cardiac actinin (C) or a-sarcomeric actin (F). In new
myocytes, dark and light blue fluorescence reflects the combination of PI and
BrdU
labeling of myocyte nuclei (C,F). Confocal microscopy; bar=10 gm.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
32
Figure 54: Effects of Time on Number and Volume of Newly Formed
Myocytes. During the interval from 10 to 20 days, developing myocytes
increased
significantly in size. However, cell number remained essentially constant. The
size
distribution was wider at 20 than at 10 days.
Figure 55: Effects of Time on the Development of Newly Formed
Coronary Vasculature. The numerical density of newly formed arterioles (Art)
and
capillaries (Cap) increased significantly during the interval from 10 to 20
days.
Results are mean SD. *P<0.05 vs 10 days.
Figure 56: Spared Myocytes in the Infarcted Ventricle. A,B, Large,
hypertrophied myocytes isolated from the remaining viable tissue of the left
ventricle and interventricular septum. Red fluorescence corresponds to cardiac
myosin antibody staining and blue fluorescence to PI labeling. Yellow
fluorescence
at the edges of the cells reflects connexin 43 (A) and N-cadherin (B).
Confocal
microscopy; bar=10
Figure 57: Cell Implantation and Echocardiography. Myocardial
regeneration attenuated ventricular dilation (A), had no effect on the
thickness of the
surviving portion of the wall (B), increased the thickness of the infarcted
region of
the ventricle (C) and improved ejection fraction (D). SO=sham-operated;
MI=untreated infarcts; MI-T=treated infarcts. Results are mean SD. *P<0.05 vs
SO;
"P<0.05 vs MI.
Figure 58: Echocardiographic Tracing. Two-dimensional images and M-
mode tracings of an untreated infarcted rat (A,B) and a treated infarcted rat
(C,D).
Panels A and C correspond to baseline conditions before coronary artery
occlusion.
The reappearance of contraction is evident in panel D (arrowheads).
Figure 59: Ventricular Function and Wall Stress. Cell implantation
improved ventricular function and attenuated the increase in diastolic wall
stress
after infarction. SO=sham-operated; MI=untreated infarcts; MI-T=treated
infarcts;
LVEDP=left ventricular end-diastolic pressure; LVDP=left ventricular developed

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
33
pressure; +dP/dt=rate of pressure rise; ¨dP/dt=rate of pressure decay. Results
are
mean SD. *P<0.05 vs SO; **P<0.05 vs MI.
Figure 60: Cell Implantation in Normal Myocardium. BrdU labeled cells
obtained at P2 were injected in sham-operated rats. Twenty days later, only a
few
undifferentiated cells were identified. A,C, Green fluorescence documents BrdU
labeling of nuclei. B,D, Myocyte cytoplasm is recognized by the red
fluorescence of
a-sarcomeric actin. Nuclei are illustrated by the blue fluorescence of PI. In
injected
cells (arrowheads), bright blue fluorescence reflects the combination of PI
and BrdU
labeling (B,D). Confocal microscopy; bar=10 pm.
Figures 61 and 62. Migration and invasion assays. Results in are reported as
the mean SD. * indicates a statistical significant difference, i.e. P <
0.05, from
cells not exposed to the growth factor.
Figure 63. Matrix metalloproteinase activity assay. Digital photograph of
the resulting gel from gelatin zymography,
Figure 64. Graphs of primitive cells expressing growth factor receptors.
The distribution of c-met and IGF-1R on c-ki? s and MDR11) s cells in the
various
regions of the heart in sham-operated (SO), infarcted-treated (Treated) and
infarcted-
untreated (Untreated) mice sacrificed 7-8 hours after surgery and 2-3 hours
after the
administration of growth factors (Treated) or saline (SO; Untreated) is shown.
These measurements include all c-kitP s and MDR1P s cells, independently of
ongoing apoptosis. Abbreviations used are as follows: A, atria; LV, left
ventricle;
R, viable myocardium remote from the infarct; B, viable myocardium bordering
the
infarct; I, non-viable infarcted myocardium. All results are reported as the
mean
SD.
Figure 65. Graphs showing the location of cycling primitive cells. The
percentage of viable Ki67 labeled c-kitP s and MDR1P s cells in the various
regions
of the heart in sham-operated (SO), infarcted-treated (Treated) and infarcted-
untreated (Untreated) mice sacrificed 7-8 hours after surgery and 2-3 hours
after the
administration of growth factors (Treated) or saline (SO; Untreated) is
presented.
Abbreviations used are as follows: A, atria; LV, left ventricle; R, viable
myocardium

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
34
remote from the infarct; B, viable myocardium bordering the infarct; I, non-
viable
infarcted myocardium. Results presented are means SD.
Figure 66. Graphs showing the frequency distribution of DNA content in
non-cycling (solid line) and cycling (broken line; Ki67 positive nuclei)
myocytes.
Both new and old myocytes showed an amount of chromatin corresponding to 2n
chromosomes. A DNA content greater than 2n was restricted to cycling nuclei.
The
measured non-cycling nuclei displayed a fluorescence intensity comparable to
that
of diploid lymphocytes. Sampling included 600 new myocytes, 1,000 old myocytes
and 1,000 lymphocytes.
Figure 67. Graphs showing the effects of myocardial infarction on the
anatomy of the heart and diastolic load. Results are presented as the mean SD.
*'**
indicate a value of p<0.05 vs. sham-operated mice (SO) and untreated infarcted
mice
(MI). MI-T refers to treated infarcted mice.
Figures 68. Graph showing the frequency distribution of myocyte sizes.
The volume of newly generated myocytes was measured in sections stained with
desmin and laminin antibodies and PI. Only longitudinally oriented cells with
centrally located nuclei were included. The length and diameter across the
nucleus
were collected in each myocyte to compute cell volume, assuming a cylindrical
shape. Four hundred cells were measured in each heart.
Figure 69: Graph showing cardiac repair. On the basis of the volume of LV
in sham-operated (SO) mice and infarct size, 42% in untreated mice (MI) and
67%
in treated mice (MI-T), the volume of myocardium destined to remain (R) and
destined to be lost (L) was computed in the two groups of infarcted mice (Fig.
9).
The volume of newly formed myocardium (F) was measured quantitatively in
treated mice. Myocardial regeneration increased the volume of remaining
myocardium (R+F) and decreased the volume of lost myocardium (L-F) by the same
amount. Therefore, infarct size in treated mice was reduced by 15%.
DETAILED DESCRIPTION
The present invention provides methods and/or pharmaceutical composition
comprising a therapeutically effective amount of somatic stem cells alone or
in
combination with a cytokine selected from the group consisting of stem cell
factor

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
(SCF), granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage
colony stimulating factor (GM-CSF), stromal cell-derived factor-1, steel
factor,
vascular endothelial growth factor, macrophage colony stimulating factor,
granulocyte-macrophage stimulating factor, hepatocyte growth factor (HGF),
5 insulin-like growth factor (IGF-1) or Interleukin-3 or any cytokine
capable of the
stimulating and/or mobilizing stem cells. Cytokines may be administered alone
or
in combination or with any other cytokine or pharmaceutical agent capable of:
the
stimulation and/or mobilization of stem cells; the maintenance of early and
late
hematopoiesis (see below); the activation of monocytes (see below),
10 macrophage/monocyte proliferation; differentiation, motility and
survival (see
below); treatment of cardiac or vascular conditions; and a pharmaceutically
acceptable carrier, diluent or excipient (including combinations thereof).
The invention also provides methods and/or pharmaceutical compositions
comprising a therapeutically effective amount of one or more cytokines for
causing
15 the migration and/or proliferation of cardiac stem cells or cardiac
primative cells
into circulatory tissue or muscle tissue or circulatory muscle tissue, e.g.,
cardiac
tissue, such as the heart or blood vessels ¨ e.g., veins, arteries, that go to
or come
from the heart such as veins and arteries directly connected or attached or
flowing
into the heart, for instance the aorta.
20 In a preferred aspect, the methods and/or compositions, including
pharmaceutical compositions, comprise effective amounts of two or more
cytokines.
More specifically, the methods and/or compositions preferably comprise
effective
amounts of hepatocyte growth factor and insulin-like growth factor-1.
The cytokines in the pharmaceutical composition of the present invention
25 may also include mediators known to be involved in the maintenance of
early and
late hematopoiesis such as IL-1 alpha and IL- 1 beta, IL-6, IL-7, IL-8, IL-11
and
IL-13; colony-stimulating factors, thrombopoietin, erythropoietin, stem cell
factor,
fit 3-ligand, hepatocyte cell growth factor, tumor necrosis factor alpha,
leukemia
inhibitory factor, transforming growth factors beta 1 and beta 3; and
macrophage
30 inflammatory protein 1 alpha), angiogenic factors (fibroblast growth
factors 1 and 2,
vascular endothelial growth factor) and mediators whose usual target (and
source) is
the connective tissue-forming cells (platelet-derived growth factor A,
epidermal
growth factor, transforming growth factors alpha and beta 2, oncostatin M and

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
36
insulin-like growth factor-1), or neuronal cells (nerve growth factor)
(Sensebe, L., et
al., Stern Cells 1997; 15:133-43), VEGF polypeptides that are present in
platelets
and megacaryocytes (Wartiovaara, U., et al., Thromb Haemost 1998; 80:171-5;
Mohle, R., Proc Natl Acad Sci USA 1997; 94:663-8) HIF-1, a potent
transcription
factor that binds to and stimulates the promoter of several genes involved in
responses to hypoxia, endothelial PAS domain protein 1 (EPAS 1),
monocyte-derived cytoldnes for enhancing collateral function such as monocyte
chemotactic protein-1 (MCP-1).
In an additionally preferred aspect, the methods and/or compositions,
including pharmaceutical compositions, comprise effective amounts of two or
more
cytokines in combination with an appropriate pharmaceutical agent useful in
treating
cardiac and/or vascular conditions.
In a preferred aspect, the pharmaceutical composition of the present
invention is delivered via injection. These routes for administration
(delivery)
include, but are not limited to subcutaneous or parenteral including
intravenous,
intraarterial, intramuscular, intraperitoneal, intramyocardial,
transendocardial, trans-
epicardial, intranasal administration as well as intrathecal, and infusion
techniques.
Hence, preferably the pharmaceutical composition is in a form that is suitable
for
injection.
When administering a therapeutic of the present invention parenterally, it
will generally be formulated in a unit dosage injectable form (solution,
suspension,
emulsion). The pharmaceutical formulations suitable for injection include
sterile
aqueous solutions or dispersions and sterile powders for reconstitution into
sterile
injectable solutions or dispersions. The carrier can be a solvent or
dispersing
medium containing, for example, water, ethanol, polyol (for example, glycerol,
propylene glycol, liquid polyethylene glycol, and the like) , suitable
mixtures
thereof, and vegetable oils.
Proper fluidity can be maintained, for example, by the use of a coating such
as lecithin, by the maintenance of the required particle size in the case of
dispersion
and by the use of surfactants. Nonaqueous vehicles such a cottonseed oil,
sesame
oil, olive oil, soybean oil, corn oil, sunflower oil, or peanut oil and
esters, such as
isopropyl myristate, may also be used as solvent systems for compound
compositions

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
37
Additionally, various additives which enhance the stability, sterility, and
isotonicity of the compositions, including antimicrobial preservatives,
antioxidants,
chelating agents, and buffers, can be added. Prevention of the action of
microorganisms can be ensured by various antibacterial and antifungal agents,
for
example, parabens, chlorobutanol, phenol, sorbic acid, and the like. In many
cases, it
will be desirable to include isotonic agents, for example, sugars, sodium
chloride,
and the like. Prolonged absorption of the injectable pharmaceutical form can
be
brought about by the use of agents delaying absorption, for example, aluminum
monostearate and gelatin. According to the present invention, however, any
vehicle,
diluent, or additive used would have to be compatible with the compounds.
Sterile injectable solutions can be prepared by incorporating the compounds
utilized in practicing the present invention in the required amount of the
appropriate
solvent with various amounts of the other ingredients, as desired.
The pharmaceutical composition of the present invention, e.g., comprising a
therapeutic compound, can be administered to the patient in an injectable
formulation containing any compatible carrier, such as various vehicles,
adjuvants,
additives, and diluents; or the compounds utilized in the present invention
can be
administered parenterally to the patient in the form of slow-release
subcutaneous
implants or targeted delivery systems such as monoclonal antibodies,
iontophoretic,
polymer matrices, liposomes, and microspheres.
The pharmaceutical composition utilized in the present invention can be
administered orally to the patient. Conventional methods such as administering
the
compounds in tablets, suspensions, solutions, emulsions, capsules, powders,
syrups
and the like are usable. Known techniques which deliver the compound orally or
intravenously and retain the biological activity are preferred.
In one embodiment, a composition of the present invention can be
administered initially, and thereafter maintained by further administration.
For
instance, a composition of the invention can be administered in one type of
composition and thereafter further administered in a different or the same
type of
composition. For example, a composition of the invention can be administered
by
intravenous injection to bring blood levels to a suitable level. The patient's
levels
are then maintained by an oral dosage form, although other forms of
administration,
dependent upon the patient's condition, can be used.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
38
It is noted that humans are treated generally longer than the mice or other
experimental animals which treatment has a length proportional to the length
of the
disease process and drug effectiveness. The doses may be single doses or
multiple
doses over a period of several days, but single doses are preferred. Thus, one
can
scale up from animal experiments, e.g., rats, mice, and the like, to humans,
by
techniques from this disclosure and documents cited herein and the knowledge
in the
art, without undue experimentation.
The treatment generally has a length proportional to the length of the disease
process and drug effectiveness and the patient being treated.
The quantity of the pharmaceutical composition to be administered will vary
for the patient being treated. In a preferred embodiment, 2 x 104¨ 1 x 105
stem cells
and 50-500 ,g/kg per day of a cytokine were administered to the patient.
While
there would be an obvious size difference between the hearts of a mouse and a
human, it is possible that 2 x 104¨ 1 x 105 stem cells would be sufficient in
a human
as well. However, the precise determination of what would be considered an
effective dose may be based on factors individual to each patient, including
their
size, age, size of the infarct, and amount of time since damage. Therefore,
dosages
can be readily ascertained by those skilled in the art from this disclosure
and the
knowledge in the art. Thus, the skilled artisan can readily determine the
amount of
compound and optional additives, vehicles, and/or carrier in compositions and
to be
administered in methods of the invention. Typically, any additives (in
addition to
the active stem cell(s) and/or cytokine(s)) are present in an amount of 0.001
to 50
wt% solution in phosphate buffered saline, and the active ingredient is
present in the
order of micrograms to milligrams, such as about 0.0001 to about 5 wt%,
preferably
about 0.0001 to about 1 wt%, most preferably about 0.0001 to about 0.05 wt% or
about 0.001 to about 20 wt%, preferably about 0.01 to about 10 wt%, and most
preferably about 0.05 to about 5 wt%. Of course, for any composition to be
administered to an animal or human, and for any particular method of
administration, it is preferred to determine therefore: toxicity, such as by
determining the lethal dose (LD) and LD50 in a suitable animal model e.g.,
rodent
such as mouse; and, the dosage of the composition(s), concentration of
components
therein and timing of administering the composition(s), which elicit a
suitable

CA 02488346 2011-06-02
39
response. Such determinations do not require undue experimentation from the
=
knowledge of the skilled artisan, this disclosure and the documents cited
herein.
And, the time for sequential administrations can be ascertained without undue
experimentation.
Additionally, one of skill in the art would be able to ascertain without undue
experimentation the appropriate pharmaceutical agent to be used in combination
with one or more cytokines; and, one of skill in the art would be able to make
the
precise determination of what would be considered an effective dose based on
factors individual to each patient, including their size, age, size of the
infarct, and
amount of time since damage. Therefore, dosages can be readily ascertained by
those skilled in the art from this disclosure and the knowledge in the art.
Examples of compositions comprising a therapeutic of the invention include
liquid preparations for orifice, e.g., oral, nasal, anal, vaginal, peroral,
intragastric,
mucosal (e.g., perlingual, alveolar, gingival, olfactory or respiratory
mucosa) etc.,
administration such as suspensions, syrups or elixirs; and, preparations for
parenteral, subcutaneous, intradennal, intramuscular or intravenous
administration
(e.g., injectable administration), such as sterile suspensions or emulsions.
Such
compositions may be in admixture with a suitable carrier, diluent, or
excipient such
as sterile water, physiological saline, glucose or the like. The compositions
can also
be lyophilized. The compositions can contain auxiliary substances such as
wetting
or emulsifying agents, pH buffering agents, gelling or viscosity enhancing
additives,
preservatives, flavoring agents, colors, an. d the like, depending upon the
route of
administration and the preparation desired. Standard texts, such as
"REMLNGTON'S PHARMACEUTICAL SCIENCE", 17th edition, 1985,
may be consulted to prepare suitable preparations,
without undue experimentation.
Compositions of the invention, are conveniently provided as liquid
preparations, e.g., isotonic aqueous solutions, suspensions, emulsions or
viscous
compositions which may be buffered to a selected pH. If digestive tract
absorption
is preferred, compositions of the invention can be in the "solid" form of
pills, tablets,
capsules, caplets and the like, including "solid" preparations which are time-
released
or which have a liquid filling, e.g., gelatin covered liquid, whereby the
gelatin is
dissolved in the stomach for delivery to the gut. If nasal or respiratory
(mucosal)

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
administration is desired, compositions may be in a form and dispensed by a
squeeze
spray dispenser, pump dispenser or aerosol dispenser. Aerosols are usually
under
pressure by means of a hydrocarbon. Pump dispensers can preferably dispense a
metered dose or, a dose having a particular particle size.
5 Compositions of the invention can contain pharmaceutically acceptable
flavors and/or colors for rendering them more appealing, especially if they
are
administered orally. The viscous compositions may be in the form of gels,
lotions,
ointments, creams and the like (e.g., for transderrnal administration) and
will
typically contain a sufficient amount of a thickening agent so that the
viscosity is
10 from about 2500 to 6500 cps, although more viscous compositions, even up
to
10,000 cps may be employed. Viscous compositions have a viscosity preferably
of
2500 to 5000 cps, since above that range they become more difficult to
administer.
However, above that range, the compositions can approach solid or gelatin
forms
which are then easily administered as a swallowed pill for oral ingestion.
15 Liquid preparations are normally easier to prepare than gels, other
viscous
compositions, and solid compositions. Additionally, liquid compositions are
somewhat more convenient to administer, especially by injection or orally.
Viscous
compositions, on the other hand, can be formulated within the appropriate
viscosity
range to provide longer contact periods with mucosa, such as the lining of the
20 stomach or nasal mucosa.
Obviously, the choice of suitable carriers and other additives will depend on
the exact route of administration and the nature of the particular dosage
form, e.g.,
liquid dosage form (e.g., whether the composition is to be formulated into a
solution,
a suspension, gel or another liquid form), or solid dosage form (e.g., whether
the
25 composition is to be formulated into a pill, tablet, capsule, caplet,
time release form
or liquid-filled form).
Solutions, suspensions and gels normally contain a major amount of water
(preferably purified water) in addition to the active compound. Minor amounts
of
other ingredients such as pH adjusters (e.g., a base such as NaOH),
emulsifiers or
30 dispersing agents, buffering agents, preservatives, wetting agents,
jelling agents,
(e.g., methylcellulose), colors and/or flavors may also be present. The
compositions
can be isotonic, i.e., they can have the same osmotic pressure as blood and
lacrimal
fluid.

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
41
The desired isotonicity of the compositions of this invention may be
accomplished using sodium chloride, or other pharmaceutically acceptable
agents
such as dextrose, boric acid, sodium tartrate, propylene glycol or other
inorganic or
organic solutes. Sodium chloride is preferred particularly for buffers
containing
sodium ions.
Viscosity of the compositions may be maintained at the selected level using a
pharmaceutically acceptable thickening agent. Methylcellulose is preferred
because
it is readily and economically available and is easy to work with. Other
suitable
thickening agents include, for example, xanthan gum, carboxymethyl cellulose,
hydroxypropyl cellulose, carbomer, and the like. The preferred concentration
of the
thickener will depend upon the agent selected. The important point is to use
an
amount which will achieve the selected viscosity. Viscous compositions are
normally prepared from solutions by the addition of such thickening agents.
A pharmaceutically acceptable preservative can be employed to increase the
shelf-life of the compositions. Benzyl alcohol may be suitable, although a
variety of
preservatives including, for example, parabens, thimerosal, chlorobutanol, or
benzalkonium chloride may also be employed. A suitable concentration of the
preservative will be from 0.02% to 2% based on the total weight although there
may
be appreciable variation depending upon the agent selected.
Those skilled in the art will recognize that the components of the
compositions should be selected to be chemically inert with respect to the
active
compound. This will present no problem to those skilled in chemical and
pharmaceutical principles, or problems can be readily avoided by reference to
standard texts or by simple experiments (not involving undue experimentation),
from this disclosure and the documents cited herein.
The inventive compositions of this invention are prepared by mixing the
ingredients following generally accepted procedures. For example the selected
components may be simply mixed in a blender, or other standard device to
produce a
concentrated mixture which may then be adjusted to the final concentration and
viscosity by the addition of water or thickening agent and possibly a buffer
to
control pH or an additional solute to control tonicity. Generally the pH may
be from
about 3 to 7.5. Compositions can be administered in dosages and by techniques
well
known to those skilled in the medical and veterinary arts taking into
consideration

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
42
such factors as the age, sex, weight, and condition of the particular patient,
and the
composition form used for administration (e.g., solid vs. liquid). Dosages for
humans or other mammals can be determined without undue experimentation by the
skilled artisan, from this disclosure, the documents cited herein, and the
knowledge
in the art.
Suitable regimes for initial administration and further doses or for
sequential
administrations also are variable, may include an initial administration
followed by
subsequent administrations; but nonetheless, may be ascertained by the skilled
artisan, from this disclosure, the documents cited herein, and the knowledge
in the
art.
The pharmaceutical compositions of the present invention are used to treat
cardiovascular diseases, including, but not limited to, atherosclerosis,
ischemia,
hypertension, restenosis, angina pectoris, rheumatic heart disease, congenital
cardiovascular defects and arterial inflammation and other diseases of the
arteries,
arterioles and capillaries or related complaint. Accordingly, the invention
involves
the administration of stem cells as herein discussed, alone or in combination
with
one or more cytokine, as herein discussed, for the treatment or prevention of
any one
or more of these conditions or other conditions involving weakness in the
heart, as
well as compositions for such treatment or prevention, use of stem cells as
herein
discussed, alone or in combination with one or more cytokine, as herein
discussed,
for formulating such compositions, and kits involving stem cells as herein
discussed,
alone or in combination with one or more cytokine, as herein discussed, for
preparing such compositions and/or for such treatment, or prevention. And,
advantageous routes of administration involves those best suited for treating
these
conditions, such as via injection, including, but are not limited to
subcutaneous or
parenteral including intravenous, intraarterial, intramuscular,
intraperitoneal,
intramyocardial, transendocardial, trans-epicardial, intranasal administration
as well
as intrathecal, and infusion techniques.
The pharmaceutical compositions of the present invention may be used as
therapeutic agents ¨ i.e. in therapy applications. As herein, the terms
"treatment" and
"therapy" include curative effects, alleviation effects, and prophylactic
effects.
As used herein, "patient" may encompass an vertebrate including but not
limited to humans, mammals, reptiles, amphibians and fish. However,

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
43
advantageously, the patient is a mammal such as a human, or an animal mammal
such as a domesticated mammal, e.g., dog, cat, horse, and the like, or
production
mammal, e.g., cow, sheep, pig, and the like
As used herein "somatic stem cell" or "stem cell" or "hematopoietic cell"
refers to either autologous or allogenic stem cells, which may be obtained
from the
bone marrow, peripheral blood, or other source.
As used herein, "adult" stem cells refers to stem cells that are not embryonic
in origin nor derived from embryos or fetal tissue.
As used herein "recently damaged myocardium" refers to myocardium which
has been damaged within one week of treatment being started. In a preferred
embodiment, the myocardium has been damaged within three days of the start of
treatment. In a further preferred embodiment, the myocardium has been damaged
within 12 hours of the start of treatment. It is advantageous to employ stem
cells
alone or in combination with cytokine(s) as herein disclosed to a recently
damaged
myocardium.
As used herein "damaged myocardium" refers to myocardial cells which
have been exposed to ischemic conditions. These ischemic conditions may be
caused by a myocardial infarction, or other cardiovascular disease or related
complaint. The lack of oxygen causes the death of the cells in the surrounding
area,
leaving an infarct, which will eventually scar.
As used herein, "home" refers to the attraction and mobilization of somatic
stem cells towards damaged myocardium and/or myocardial cells.
As used herein, "assemble" refers to the assembly of differentiated somatic
stem cells into functional structures i.e., myocardium and/or myocardial
cells,
coronary arteries, arterioles, and capillaries etc. This assembly provides
functionality to the differentiated myocardium and/or myocardial cells,
coronary
arteries, arterioles and capillaries.
Thus, the invention involves the use of somatic stem cells. These are present
in animals in small amounts, but methods of collecting stem cells are known to
those skilled in the art.
In another aspect of the invention, the stem cells are selected to be lineage
negative. The term "lineage negative" is known to one skilled in the art as
meaning
the cell does not express antigens characteristic of specific cell lineages.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
44
Advantageously, the lineage negative stem cells are selected to be c-kit
positive. The term "c-kit" is known to one skilled in the art as being a
receptor
which is known to be present on the surface of stem cells, and which is
routinely
utilized in the process of identifying and separating stem cells from other
surrounding cells.
The invention further involves a therapeutically effective dose or amount of
stem cells applied to the heart. An effective dose is an amount sufficient to
effect a
beneficial or desired clinical result. Said dose could be administered in one
or more
administrations. In the examples that follow, 2 x 104¨ 1 x 105 stem cells were
administered in the mouse model. While there would be an obvious size
difference
between the hearts of a mouse and a human, it is possible that this range of
stem
cells would be sufficient in a human as well. However, the precise
determination of
what would be considered an effective dose may be based on factors individual
to
each patient, including their size, age, size of the infarct, and amount of
time since
damage. One skilled in the art, specifically a physician or cardiologist,
would be
able to determine the number of stem cells that would constitute an effective
dose
without undue experimentation.
In another aspect of the invention, the stem cells are delivered to the heart,
specifically to the border area of the infarct. As one skilled in the art
would be
aware, the infarcted area is visible grossly, allowing this specific placement
of stem
cells to be possible.
The stem cells are advantageously administered by injection, specifically an
intramyocardial injection. As one skilled in the art would be aware, this is
the
preferred method of delivery for stem cells as the heart is a functioning
muscle.
Injection of the stem cells into the heart ensures that they will not be lost
due to the
contracting movements of the heart.
In a further aspect of the invention, the stem cells are administered by
injection transendocardially or trans-epicardially. This preferred embodiment
allows
the stem cells to penetrate the protective surrounding membrane, necessitated
by the
embodiment in which the cells are injected intramyocardially.
A preferred embodiment of the invention includes use of a catheter-based
approach to deliver the trans-endocardial injection. The use of a catheter
precludes
more invasive methods of delivery wherein the opening of the chest cavity
would be

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
necessitated. As one skilled in the art is aware, optimum time of recovery
would be
allowed by the more minimally invasive procedure, which as outlined here,
includes
a catheter approach.
Further embodiments of the invention require the stem cells to migrate into
5 the infarcted region and differentiate into myocytes, smooth muscle
cells, and
endothelial cells. It is known in the art that these types of cells must be
present to
restore both structural and functional integrity. Other approaches to
repairing
infarcted or ischemic tissue have involved the implantation of these cells
directly
into the heart, or as cultured grafts, such as in U.S. Patent No. 6,110,459,
and
10 6,099,832.
Another embodiment of the invention includes the proliferation of the
differentiated cells and the formation of the cells into cardiac structures
including
coronary arteries, arterioles, capillaries, and myocardium. As one skilled in
the art is
aware, all of these structures are essential for proper function in the heart.
It has
15 been shown in the literature that implantation of cells including
endothelial cells and
smooth muscle cells will allow for the implanted cells to live within the
infarcted
region, however they do not form the necessary structures to enable the heart
to
regain full functionality. The ability to restore both functional and
structural
integrity is yet another aspect of this invention.
20 Another aspect of the invention relates to the administration of a
cytokine.
This cytokine may be chosen from a group of cytokines, or may include
combinations of cytokines. Stem cell factor (SCF) and granulocyte-colony
stimulating factor (G-CSF) are known by those skilled in the art as
stimulating
factors which cause the mobilization of stem cells into the blood stream
(Bianco et
25 al, 2001, Clutterbuck, 1997, Kronenwett et al, 2000, LaIuppa et al,
1997, Patchen et
al, 1998). Stromal cell-derived factor-1 has been shown to stimulate stem cell
mobilization chemotactically, while steel factor has both chemotactic and
chemoldnetic properties (Caceres-Cortes et al, 2001, Jo et al, 2000, Kim and
Broxmeyer, 1998, lkuta et al, 1991). Vascular endothelial growth factor has
been
30 surmised to engage a paracrine loop that helps facilitate migration
during
mobilization (Bautz et al, 2000, Janowska-Wieczorek et al, 2001) . Macrophage
colony stimulating factor and granulocyte-macrophage stimulating factor have
been
shown to function in the same manner of SCF and G-CSF, by stimulating

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
46
mobilization of stem cells. Interleukin-3 has also been shown to stimulate
mobilization of stem cells, and is especially potent in combination with other
cytokines.
The cytokine can be administered via a vector that expresses the cytokine in
vivo. A vector for in vivo expression can be a vector or cells or an
expression
system as cited in any document incorporated herein by reference or used in
the art,
such as a viral vector, e.g., an adenovitus, poxvirus (such as vaccinia,
canarypox
virus, MVA, NYVAC, ALVAC, and the like), lentivirus or a DNA plasmid vector;
and, the cytokine can also be from in vitro expression via such a vector or
cells or
expression system or others such as a baculovirus expression system, bacterial
vectors such as E. coli, and mammalian cells such as CHO cells. See, e.g.,
U.S.
Patent Nos. 6,265,189, 6,130,066, 6,004,777, 5,990,091, 5,942,235, 5,833,975.
The
cytokine compositions may lend themselves to administration by routes outside
of
those stated to be advantageous or preferred for stem cell preparations; but,
cytokine
compositions may also be advantageously administered by routes stated to be
advantageous or preferred for stem cell preparations.
A further aspect of the invention involves administration of a therapeutically
effective dose or amount of a cytokine. An effective dose is an amount
sufficient to
effect a beneficial or desired clinical result. Said dose could be
administered in one
or more administrations. In a preferred embodiment, the dose would be given
over
the course of about two or three days following the beginning of treatment.
However, the precise determination of what would be considered an effective
dose
may be based on factors individual to each patient, including their size, age,
size of
the infarct, the cytokine or combination of cytokines being administered, and
amount of time since damage. One skilled in the art, specifically a physician
or
cardiologist, would be able to determine a sufficient amount of cytokine that
would
constitute an effective dose without being subjected to undue experimentation.
The invention also involves the administration of the therapeutically
effective dose or amount of a cytokine being delivered by injection,
specifically
subcutaneously or intravenously. A person skilled in the art will be aware
that
subcutaneous injection or intravenous delivery are extremely common and offer
an
effective method of delivering the specific dose in a manner which allows for
timely
uptake and circulation in the blood stream.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
47
A further aspect of the invention includes the administered cytokine
stimulating the patient's stem cells and causing mobilization into the blood
stream.
As mentioned previously, the given cytokines are well-known to one skilled in
the
art for their ability to promote said mobilization.
Advantageously, once the stem cells have mobilized into the bloodstream,
they home to the damaged area of the heart, as will become clear through the
following examples.
Further embodiments of the invention involve the stem cells migrating into
the infarcted region and differentiating into myocytes, smooth muscle cells,
and
endothelial cells. It is known in the art that these types of cells must be
present to
restore both structural and functional integrity.
A further embodiment of the invention includes administering an effective
amount of one or more cytokines to the infarcted region. An effective dose is
an
amount sufficient to effect a beneficial or desired clinical result. Said dose
could be
administered in one or more administrations. However, the precise
determination of
what would be considered an effective dose may be based on factors individual
to
each patient, including their size, age, size of the infarct, the cytokine or
combination of cytokines being administered, and amount of time since damage.
One skilled in the art, specifically a physician or cardiologist, would be
able to
determine a sufficient amount of cytokine that would constitute an effective
dose
without being subjected to undue experimentation.
A still further embodiment of the invention includes the administering of an
effective amount of one or more cytokines to the heart by injection.
Preferably, the
cytokines are delivered to the infarcted region or to the area bordering the
infarcted
region. As one skilled in the art would be aware, the infarcted area is
visible
grossly, allowing this specific placement of cytokines to be possible.
The cytokines are advantageously administered by injection, specifically an
intramyocardial injection. As one skilled in the art would be aware, this is
the
preferred method of delivery for cytokines as the heart is a functioning
muscle.
Injection of the cytokines into the heart ensures that they will not be lost
due to the
contracting movements of the heart.
In a further aspect of the invention, the cytokines are administered by
injection transendocardially or trans-epicardially. This preferred embodiment
allows

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
48
the cytokines to penetrate the protective surrounding membrane, necessitated
by the
embodiment in which the cytokines are injected intramyocardially.
A preferred embodiment of the invention includes use of a catheter-based
approach to deliver the trans-endocardial injection. The use of a catheter
precludes
more invasive methods of delivery wherein the opening of the chest cavity
would be
necessitated. As one skilled in the art is aware, optimum time of recovery
would be
allowed by the more minimally invasive procedure, which as outlined here,
includes
a catheter approach.
A further embodiment of the invention includes the delivery of the cytokines
by a single administration. A still further embodiment of the invention
includes
multiple administrations of the same dosage of cytokines to the heart. A still
further
embodiment of the invention includes administration of multiple doses of the
cytokines to the heart, such that a gradient is fowled.
A still further embodiment of the invention includes the stimulation,
migration, proliferation and/or differentiation of the resident cardiac stem
cells.
Another embodiment of the invention includes the proliferation of the
differentiated cells and the formation of the cells into cardiac structures
including
coronary arteries, arterioles, capillaries, and myocardium. As one skilled in
the art is
aware, all of these structures are important for proper function in the heart.
It has
been shown in the literature that implantation of cells including endothelial
cells and
smooth muscle cells will allow for the implanted cells to live within the
infarcted
region, however they do not form the necessary structures to enable the heart
to
regain full functionality. The ability to restore both functional and
structural
integrity or better functional and structural integrity than previously
achieved in the
art is yet another aspect of this invention.
It is a preferred in the practice of the invention to utilize both the
administration of stem cells and that of a cytokine to ensure the most
effective
method of repairing damaged myocardium.
Stem cells employed in the invention are advantageously selected to be
lineage negative. The term "lineage negative" is known to one skilled in the
art as
meaning the cell does not express antigens characteristic of specific cell
lineages.
And, it is advantageous that the lineage negative stem cells are selected to
be c-kit
positive. The term "c-kit" is known to one skilled in the art as being a
receptor

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
49
which is known to be present on the surface of stem cells, and which is
routinely
utilized in the process of identifying and separating stem cells from other
surrounding cells.
In certain embodiments, a therapeutically effective dose of stem cells is
applied, delivered, or administered to the heart or implanted into the heart.
An
effective dose or amount is an amount sufficient to effect a beneficial or
desired
clinical result. Said dose could be administered in one or more
administrations. In
the examples that follow, 2 x 104 ¨ 1 x 105 stem cells were administered in
the
mouse model. While there would be an obvious size difference between the
hearts
of a mouse and a human, it is possible that 2 x 104 ¨ 1 x 105 stem cells would
be
sufficient in a human as well. However, the precise determination of what
would be
considered an effective dose may be based on factors individual to each
patient,
including their size, age, size of the infarct, and amount of time since
damage. One
skilled in the art, specifically a physician or cardiologist, would be able to
determine
the number and type (or types) of stem cells which would constitute an
effective
dose without being subjected to undue experimentation, from this disclosure
and the
knowledge in the art; and, in this regard and in general in regard to
preparing
formulations and administering formulations or components thereof, mention is
made of the teachings in the Examples and that the skilled artisan can scale
dosages,
amounts and the like based on the weight of the patient to be treated in
comparison
to the weight of any animal employed in the Examples. The stem cells are
advantageously bone marrow or are cardiac stem cells; and even more
advantageously, the stem cells are adult bone marrow (hematopoietic stem
cells) or
adult cardiac stem cells or a combination thereof or a combination of cardiac
stem
cells such as adult cardiac stem cells and another type of stem cell such as
another
type of adult stem cells.
In another aspect of the invention, the stem cells are delivered to the heart,
specifically to the border area of the infarct. As one skilled in the art
would be
aware, the infarcted area is visible grossly, allowing this specific placement
of stem
cells to be possible.
The stem cells are advantageously administered by injection, specifically an
intramyocardial injection. As one skilled in the art would be aware, this is
the
preferred method of delivery for stem cells as the heart is a functioning
muscle.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
Injection of the stem cells into the heart ensures that they will not be lost
due to the
contracting movements of the heart.
In other aspects of the invention, the stem cells are administered by
injection
transendocardially or trans-epicardially. This preferred embodiment allows the
stem
5 cells to penetrate the protective surrounding membrane, necessitated by
the
embodiment in which the cells are injected intramyocardially.
A preferred embodiment of the invention includes use of a catheter-based
approach to deliver the trans-endocardial injection. The use of a catheter
precludes
more invasive methods of delivery wherein the opening of the chest cavity
would be
10 necessitated. As one skilled in the art is aware, optimum time of
recovery would be
allowed by the more minimally invasive procedure, which as outlined here,
includes
a catheter approach.
Embodiments of the invention can involve the administration of a cytokine.
This cytokine may be chosen from a group of cytokines, or may include
15 combinations of cytokines.
A further aspect of the invention involves administration of a therapeutically
effective dose of a cytokine. An effective dose or amount is an amount
sufficient to
effect a beneficial or desired clinical result. Said dose could be
administered in one
or more administrations. In a preferred embodiment, the dose would be given
over
20 the course of about two or three days following the beginning of
treatment.
However, the precise determination of what would be considered an effective
dose
may be based on factors individual to each patient, including their size, age,
size of
the infarct, the cytokine or combination of cytokines being administered, and
amount of time since damage. One skilled in the art, specifically a physician
or
25 cardiologist, would be able to determine a sufficient amount of cytokine
that would
constitute an effective dose without being subjected to undue experimentation,
especially in view of the disclosure herein and the knowledge in the art.
The administration of the therapeutically effective dose of at least one
cytokine is advantageously by injection, specifically subcutaneously or
30 intravenously. A person skilled in the art will be aware that
subcutenous injection or
intravenous delivery are extremely common and offer an effective method of
delivering the specific dose in a manner which allows for timely uptake and
circulation in the blood stream.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
51
A further aspect of the invention includes the administered cytokine
stimulating the patient's stem cells and causing mobilization into the blood
stream.
As mentioned previously, the given cytokines are well known to one skilled in
the
art for their ability to promote said mobilization. Again, once the stem cells
have
mobilized into the bloodstream, they home to the damaged area of the heart.
Thus in
certain embodiments, both the implanted stem cells and the mobilized stem
cells
migrate into the infarct region and differentiate into myocytes, smooth muscle
cells,
and endothelial cells. It is known in the art that these types of cells are
advantageously present to restore both structural and functional integrity.
Another embodiment of the invention includes the proliferation of the
differentiated cells and the formation of the cells into cardiac structures
including
coronary arteries, arterioles, capillaries, and myocardium. As one skilled in
the art is
aware, all of these structures are essential for proper function in the heart.
It has
been shown in the literature that implantation of cells including endothelial
cells and
smooth muscle cells will allow for the implanted cells to live within the
infarcted
region, however they do not form the necessary structures to enable the heart
to
regain full functionality. Cardiac structures can be generated ex vivo and
then
implanted in the form of a graft; with the implantation of the graft being
alone or in
combination with stem cells or stem cells and at least one cytokine as in this
disclosure, e.g., advantageously adult or cardiac or hematopoietic stem cells
such as
adult cardiac and/or adult hematpoietic stem cells or adult cardiac stem cells
with
another type of stem cell e.g. another type of adult stem cell. The means of
generating and/or regenerating myocardium ex vivo, may incorporate somatic
stem
cells and heart tissue being cultured in vitro, optionally in the presence of
a ctyokine.
The somatic stem cells differentiate into myocytes, smooth muscle cells and
endothelial cells, and proliferate in vitro, forming myocardial tissue and/or
cells.
These tissues and cells may assemble into cardiac structures including
arteries,
arterioles, capillaries, and myocardium. The tissue and/or cells formed in
vitro may
then be implanted into a patient, e.g. via a graft, to restore structural and
functional
integrity.
Additionally or alternatively, the source of the tissue being grafted can be
from other sources of tissue used in grafts of the heart.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
52
The restoration or some restoration of both functional and structural
integrity
of cardiac tissue ¨ advantageously over that which has occurred previously -
is yet
another aspect of this invention.
Accordingly, the invention comprehends, in further aspects, methods for
preparing compositions such as pharmaceutical compositions including somatic
stem cells and/or at least one cytokine, for instance, for use in inventive
methods for
treating cardiovascular disease or conditions or cardiac conditions.
The present invention is additionally described by way of the following, non-
limiting examples, that provide a better understanding of the present
invention and
of its many advantages.
All of the materials, reagents, chemicals, assays, cytokines, antibodies, and
miscellaneous items referred to in the following examples are readily
available to
the research community through commercial suppliers, including but not limited
to,
Genzyme, Invitrogen, Gibco BRL, Clonetics, Fisher Scientific, R& D Systems,
MBL International Corporation, CN Biosciences Corporate, Sigma Aldrich, and
CedarLane Laboratories, Limited.
For example,
stem cell factor is available under the name SCF (multiple forms of
recombinant human, recombinant mouse, and antibodies to each),
from R & D Systems (614 McKinley Place N.E., Minneapolis, MN
55413);
granulocyte-colony stimulating factor is available under the name G-CSF
(multiple forms of recombinant human, recombinant mouse, and
antibodies to each), from R & D Systems;
stem cell antibody-1 is available under the name SCA-1 from MBL
International Corporation (200 Dexter Avenue, Suite D, Watertown,
MA 02472);
multidrug resistant antibody is available under the name Anti-MDR from CN
Biosciences Corporate;
c-kit antibody is available under the name c-kit (Ab-1) Polyclonal Antibody
from CN Biosciences Corporate (Affiliate of Merck KgaA,

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
53
Darmstadt, Germany. Corporate headquarters located at 10394
Pacific Center Court, San Diego, CA 92121).
EXAMPLES
EXAMPLE 1: Hematopoietic Stem Cell (HS C) Repair of Infarcted Myocardium
A. Harvesting of Hematopoietic Stem Cells
Bone marrow was harvested from the femurs and tibias of male transgenic
mice expressing enhanced green fluorescent protein (EGFP). After surgical
removal
of the femurs and tibias, the muscle was dissected and the upper and lower
surface
of the bone was cut on the surface to allow the collecting buffer to
infiltrate the bone
marrow. The fluid containing buffer and cells was collected in tubes such as
1.5 ml
Epindorf tubes. Bone marrow cells were suspended in PBS containing 5% fetal
calf
serum (FCS) and incubated on ice with rat anti-mouse monoclonal antibodies
specific for the following hematopoietic lineages: CD4 and CD8 (T-
lymphocytes),
B-220 (B-lymphocytes), Mac-1 (macrophages), GR-1 (granulocytes) (Caltag
Laboratories) and TER-119 (erythrocytes) (Pharmingen). Cells were then rinsed
in
PBS and incubated for 30 minutes with magnetic beads coated with goat anti-rat
immunoglobulin (Polysciences Inc.). Lineage positive cells (Lin+) were removed
by
a biomagnet and lineage negative cells (Lin-) were stained with ACK-4-biotin
(anti-
c-kit mAb). Cells were rinsed in PBS, stained with streptavidin-conjugated
phycoerythrin (SA-PE) (Caltag Labs.) and sorted by fluorescence activated cell
sorting (FACS) using a FACS Vantage instrument (Becton Dickinson). Excitation
of
EGFP and ACK-4-biotin-SA-EP occurred at a wavelength of 488 nm. The Lin- cells
were sorted as c-kit positive (c-kitP s) and c-kit negative (c-kitNEG) with a
1-2 log
difference in staining intensity (Figure 1). The c-kitP s cells were suspended
at 2 x
104 to 1 x 105 cells in 5 ul of PBS and the c-kitNE cells were suspended at a
concentration of 1 x 105 in 5 IA of PBS.
B. Induction of Myocardial Infarction in Mice
Myocardial infarction was induced in female C57BL/6 mice at 2 months of
age as described by Li et al. (1997). Three to five hours after infarction,
the thorax
of the mice was reopened and 2.5 t1 of PBS containing Lin-c-kirs cells were
injected in the anterior and posterior aspects of the viable myocardium
bordering the

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
54
infarct (Figure 2). Infarcted mice, left uninjected or injected with Lin-c-kit-
NEG cells,
and sham-operated mice i.e., mice where the chest cavity was opened but no
infarction was induced, were used as controls. All animals were sacrificed 9
2
days after surgery. Protocols were approved by institutional review board.
Results
are presented as mean SD. Significance between two measurements was
determined by the Student's t test, and in multiple comparisons was evaluated
by the
Bonferroni method (Scholzen and Gerdes, 2000). P<0.05 was considered
significant.
Injection of male Lin- c-ki?Gs bone marrow cells in the pen-infarcted left
ventricle of female mice resulted in myocardial regeneration. The pen-
infarcted
region is the region of viable myocardium bordering the infarct. Repair was
obtained in 12 of 30 mice (40%). Failure to reconstitute infarcts was
attributed to
the difficulty of transplanting cells into tissue contracting at 600 beats per
minute
(bpm). However, an immunologic reaction to the histocompatibility antigen on
the
Y chromosome of the donor bone marrow cells could account for the lack of
repair
in some of the female recipients. Closely packed myocytes occupied 68 11% of
the
infarcted region and extended from the anterior to the posterior aspect of the
ventricle (Figs. 2A-2D). New myocytes were not found in mice injected with Lin-
c-
kitNEG cells (Fig. 2E).
C. Determination of Ventricular Function
Mice were anesthetized with chloral hydrate (400 mg/kg body weight, i.p.),
and the right carotid artery was cannulated with a microtip pressure
transducer
(model SPR-671, Millar) for the measurements of left ventricular (LV)
pressures
and LV + and ¨dP/dt in the closed-chest preparation to determine whether
developing myocytes derived from the HSC transplant had an impact on function.
Infarcted mice non-injected or injected with Lin-c-kitNEG cells were combined
in the
statistics. In comparison with sham-operated groups, the infarcted groups
exhibited
indices of cardiac failure (Figure 3). In mice treated with Lin-c-ki?Gs cells,
LV end-
diastolic pressure (LVEDP) was 36% lower, and developed pressure (LVDP) and
LV+ and ¨dP/dt were 32%, 40%, and 41% higher, respectively (Figure 4A).
D. Determination of Cell Proliferation and EGFP Detection

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
The abdominal aorta was cannulated, the heart was arrested in diastole by
injection of cadmium chloride (CdC12), and the myocardium was perfused
retrogradely with 10% buffered formalin. Three tissue sections, from the base
to the
apex of the left ventricle, were stained with hematoxylin and eosin. At 9 2
days
5 after coronary occlusion, the infarcted portion of the ventricle was
easily identifiable
grossly and histologically (see Fig. 2A). The lengths of the endocardial and
epicardial surfaces delimiting the infarcted region, and the endocardium and
epicardium of the entire left ventricle were measured in each section.
Subsequently,
their quotients were computed to yield the average infarct size in each case.
This
10 was accomplished at 4X magnification utilizing an image analyzer
connected to a
microscope. The fraction of endocardial and epicardial circumference
delimiting the
infarcted area (Pfeffer and Braunwald, 1990; Li et al., 1997) did not differ
in
untreated mice, 78 18% (n=8) and in mice treated with Lin- c-ki?Gs cells (n=-
12),
75 14% or Lin- c-kitNEG cells (n=11), 75 15%.
15 To
establish whether Lin- c-kitl'Gs cells resulted in myocardial regeneration,
BrdU (50 mg/kg body weight, i.p.) was administered daily to the animals for 4-
5
consecutive days before sacrifice to determine cumulative cell division during
active
growth. Sections were incubated with anti-BrdU antibody and BrdU labeling of
cardiac cell nuclei in the S phase was measured. Moreover, expression of Ki67
in
20 nuclei (Ki67 is expressed in cycling cells in Gl, S, G2, and early
mitosis) was
evaluated by treating samples with a rabbit polyclonal anti-mouse Ki67
antibody
(Dako Corp.). FITC-conjugated goat anti-rabbit IgG was used as secondary
antibody. (Figure 5 and 6). EGFP was detected with a rabbit polyclonal anti-
GFP
(Molecular Probes). Myocytes were recognized with a mouse monoclonal anti-
25 cardiac myosin heavy chain (MAB 1548; Chemicon) or a mouse monoclonal
anti-a-
sarcomeric actin (clone 5C5; Sigma), endothelial cells with a rabbit
polyclonal anti-
human factor VIII (Sigma) and smooth muscle cells with a mouse monoclonal anti-
a-smooth muscle actin (clone 1A4; Sigma). Nuclei were stained with propidium
iodide (PI), 10 ,g/ml. The percentages of myocyte (M), endothelial cell (EC)
and
30 smooth muscle cell (SMC) nuclei labeled by BrdU and Ki67 were obtained
by
confocal microscopy. This was accomplished by dividing the number of nuclei
labeled by the total number of nuclei examined. Number of nuclei sampled in
each

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
56
cell population was as follows; BrdU labeling: M=2,908; EC=2,153; SMC=4,877.
Ki67 labeling: M=3,771; EC=4,051; SMC=4,752. Number of cells counted for
EGFP labeling: M=3,278; EC=2,056; SMC=1,274! The percentage of myocytes in
the regenerating myocardium was determined by delineating the area occupied by
cardiac myosin stained cells divided by the total area represented by the
infarcted
region in each case. Myocyte proliferation was 93% (p<0.001) and 60% (p<0.001)
higher than in endothelial cells, and 225% (p<0.001 and 176% (p<0.001) higher
than smooth muscle cells, when measured by BrdU and Ki67, respectively.
The origin of the cells in the forming myocardium was determined by the
expression of EGFP (Fig. 7 and 8). EGFP expression was restricted to the
cytoplasm and the Y chromosome to nuclei of new cardiac cells. EGFP was
combined with labeling of proteins specific for myocytes, endothelial cells
and
smooth muscle cells. This allowed the identification of each cardiac cell type
and
the recognition of endothelial cells and smooth muscle cells organized in
coronary
vessels (Figs. 5, 7, and 8). The percentage of new myocytes, endothelial cells
and
smooth muscle cells that expressed EGFP was 53 9% (n=7), 44 6% (n=7) and
49 7% (n=7), respectively. These values were consistent with the fraction of
transplanted Lin- c-ki? s bone marrow cells that expressed EGFP, 44 10% (n=6).
An average 54 8% (n=6) of myocytes, endothelial cells and smooth muscle cells
expressed EGFP in the heart of donor transgenic mice.
E. Detection of the Y-Chromosome
For the fluorescence in situ hybridization (FISH) assay, sections were
exposed to a denaturing solution containing 70% formamide. After dehydration
with ethanol, sections were hybridized with the DNA probe CEP Y (satellite
III)
Spectrum Green (Vysis) for 3 hours. Nuclei were stained with PI.
Y-chromosomes were not detected in cells from the surviving portion of the
ventricle. However, the Y-chromosome was detected in the newly formed
myocytes, indicating their origin as from the injected bone marrow cells (Fig.
9).
F. Detection of Transription Factors and Connexin 43
Sections were incubated with rabbit polyclonal anti-MEF2 (C-21; Santa
Cruz), rabbit polyclonal anti-GATA-4 (H-112; Santa Cruz), rabbit polyclonal
anti-
Csx/Nloc2.5 (obtained from Dr. Izumo) and rabbit polyclonal anti-connexin 43

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
57
(Sigma). FITC-conjugated goat anti-rabbit IgG (Sigma) was used as secondary
antibody.
To confirm that newly formed myocytes represented maturing cells aiming at
functional competence, the expression of the myocyte enhancer factor 2 (MEF2),
the
cardiac specific transcription factor GATA-4 and the early marker of myocyte
development Csx/Nkx2.5 was examined. In the heart, MEF2 proteins are recruited
by GATA-4 to synergistically activate the promoters of several cardiac genes
such
as myosin light chain, troponin T, troponin I, a-myosin heavy chain, desmin,
atrial
natriuretic factor and a-actin (Durocher et al., 1997; Morin et al., 2000).
Csx/Nkx2.5 is a transcription factor restricted to the initial phases of
myocyte
differentiation (Durocher et al., 1997). In the reconstituting heart, all
nuclei of
cardiac myosin labeled cells expressed MEF2 (Figs. 7D-7F) and GATA-4 (Fig.
10),
but only 4019% expressed Csx/Nkx2.5 (Figs. 7G-7I). To characterize further the
properties of these myocytes, the expression of connexin 43 was determined.
This
protein is responsible for intercellular connections and electrical coupling
through
the generation of plasma membrane channels between myocytes (Beardsle et al.,
1998; Musil et al., 2000); connexin 43 was apparent in the cell cytoplasm and
at the
surface of closely aligned differentiating cells (Figs. 11A-11D). These
results were
consistent with the expected functional competence of the heart muscle
phenotype.
Additionally, myocytes at various stages of maturation were detected within
the
same and different bands (Fig. 12).
EXAMPLE 2: Mobilization of Bone Marrow Cells to Repair Infarcted Myocardium
A. Myocardial Infarction and Cytokines.
Fifteen C57BL/6 male mice at 2 months of age were splenectomized and 2
weeks later were injected subcutaneously with recombinant rat stem cell factor
(SCF), 200 g/kg/day, and recombinant human granulocyte colony stimulating
factor (G-CSF), 50 jig/kg/day (Amgen), once a day for 5 days (Bodine et al.,
1994;
Orlic et al., 1993). Under ether anesthesia, the left ventricle (LV) was
exposed and
the coronary artery was ligated (Orlic et al., 2001; Li et al., 1997; Li et
al., 1999).
SCF and G-CSF were given for 3 more days. Controls consisted of splenectomized
infarcted and sham-operated (SO) mice injected with saline. BrdU, 50 mg/kg
body

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
58
weight, was given once a day, for 13 days, before sacrifice; mice were killed
at 27
days. Protocols were approved by New York Medical College. Results are
mean SD. Significance was determined by the Student's t test and Bonferroni
method (Li et al., 1999). Mortality was computed with log-rank test. P<0.05
was
significant.
Given the ability of bone marrow Lin- c-kitP s cells to transdifferentiate
into
the cardiogenic lineage (Orlic et al., 2001), a protocol was used to maximize
their
number in the peripheral circulation in order to increase the probability of
their
homing to the region of dead myocardium. In normal animals, the frequency of
Lin-
c-kirs cells in the blood is only a small fraction of similar cells present in
the bone
marrow (Bodine et al., 1994; Orlic et al., 1993). As documented previously,
the
cytokine treatment used here promotes a marked increase of Lin- c-kitP s cells
in the
bone marrow and a redisiribution of these cells from the bone marrow to the
peripheral blood. This protocol leads to a 250-fold increase in Lin- c-kit' s
cells in
the circulation (Bodine et al., 1994; Orlic et al., 1993).
In the current study, BMC mobilization by SCF and G-CSF resulted in a
dramatic increase in survival of infarcted mice; with cytokine treatment, 73%
of
mice (11 of 15) survived 27 days, while mortality was very high in untreated
infarcted mice (Fig. 13A). A large number of animals in this group died from 3
to 6
days after myocardial infarction (MI) and only 17% (9 of 52) reached 27 days
(p<0.001). Mice that died within 48 hours post-MI were not included in the
mortality curve to minimize the influence of the surgical trauma. Infarct size
was
similar in the cytokine-, 64 11% (n=11), and saline-, 62 9% (n=9), injected
animals
as measured by the number of myocytes lost in the left ventricular free wall
(LVFW)
at 27 days (Fig. 14).
Importantly, bone marrow cell mobilization promoted myocardial
regeneration in all 11 cytokine-treated infarcted mice, sacrificed 27 days
after
surgery (Fig. 13B). Myocardial growth within the infarct was also seen in the
4
mice that died prematurely at day 6 (n=2) and at day 9 (n=2). Cardiac repair
was
characterized by a band of newly formed myocardium occupying most of the
damaged area. The developing tissue extended from the border zone to the
inside of
the injured region and from the endocardium to the epicardium of the LVFW. In
the
absence of cytokines, myocardial replacement was never observed and healing
with

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
59
scar formation was apparent (Fig. 13C). Conversely, only small areas of
collagen
accumulation were detected in treated mice.
B. Detection of BMC Mobilization by Echocardiography and
Hemodynamics.
Echocardiography was performed in conscious mice using a Sequoia 256c
(Acuson) equipped with a 13-MHz linear transducer (15L8). The anterior chest
area
was shaved and two dimensional (2D) images and M-mode tracings were recorded
from the parasternal short axis view at the level of papillary muscles. From M-
mode
tracings, anatomical parameters in diastole and systole were obtained (Pollick
et al.,
1995). Ejection fraction (EF) was derived from LV cross sectional area in 2D
short
axis view (Pollick et al., 1995): EF= [(LVDA-LVSA)/LVDA]* 100 where LVDA
and LVSA correspond to LV areas in diastole and in systole. Mice were
anesthetized with chloral hydrate (400 mg/kg body weight, ip) and a microtip
pressure transducer (SPR-671, Millar) connected to a chart recorder was
advanced
into the LV for the evaluation of pressures and + and - dP/dt in the closed-
chest
preparation (Orlic et al., 2001; Li et al., 1997; Li et al., 1999).
EF was 48%, 62% and 114% higher in treated than in non-treated mice at 9,
16 and 26 days after coronary occlusion, respectively (Fig. 15D). In mice
exposed
to cytokines, contractile function developed with time in the infarcted region
of the
wall (Figs. 15E-M; Figs. 16H-P, www.pnas.org). Conversely, LV end-diastolic
pressure (LVEDP) increased 76% more in non-treated mice. The changes in LV
systolic pressure (not shown), developed pressure (LVDP), + and -dP/dt were
also
more severe in the absence of cytokine treatment (Figs. 17A-D). Additionally,
the
increase in diastolic stress in the zone bordering and remote from infarction
was 69-
73% lower in cytokine-treated mice (Fig. 15N). Therefore, cytokine-mediated
infarct repair restored a noticeable level of contraction in the regenerating
myocardium, decreasing diastolic wall stress and increasing ventricular
performance. Myocardial regeneration attenuated cavitary dilation and mural
thinning during the evolution of the infarcted heart in vivo.
Echocardiographically, LV end-systolic (LVESD) and end-diastolic
(LVEDD) diameters increased more in non-treated than in cytokine-treated mice,
at
9, 16 and 26 days after infarction (Figs. 16A-B). Infarction prevented the
evaluation
of systolic (AWST) and diastolic (AWDT) anterior wall thickness. When

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
measurable, the posterior wall thickness in systole (PWST) and diastole (PWDT)
was greater in treated mice (Figs. 16C-D). Anatomically, the wall bordering
and
remote from infarction was 26% and 22% thicker in cytokine-injected mice (Fig.
16E). BMC-induced repair resulted in a 42% higher wall thickness-to-chamber
5 radius ratio (Fig. 15A). Additionally, tissue regeneration decreased the
expansion in
cavitary diameter, -14%, longitudinal axis, -5% (Figs.16F-G), and chamber
volume,-
26% (Fig. 15B). Importantly, ventricular mass-to-chamber volume ratio was 36%
higher in treated animals (Fig. 15C). Therefore, BMC mobilization that led to
proliferation and differentiation of a new population of myocytes and vascular
10 structures attenuated the anatomical variables which define cardiac
decompensation.
C. Cardiac Anatomy and Deteimination of Infarct Size.
Following hemodynamic measurements, the abdominal aorta was
cannulated, the heart was arrested in diastole with CdC12 and the myocardium
was
perfused with 10% formalin. The LV chamber was filled with fixative at a
pressure
15 equal to the in vivo measured end-diastolic pressure (Li et al., 1997;
Li et al., 1999).
The LV intracavitary axis was measured and three transverse slices from the
base,
mid-region and apex were embedded in paraffin. The mid-section was used to
measure LV thickness, chamber diameter and volume (Li et al., 1997; Li et al.,
1999). Infarct size was deteanined by the number of myocytes lost from the
LVFW
20 (Olivetti et al., 1991; Beltrami et al., 1994).
To quantify the contribution of the developing band to the ventricular mass,
firstly the volume of the LVFW (weight divided by 1.06 g/m1) was determined in
each group of mice. The data was 56 2 mm3 in sham operated (SO), 62 4 mm3
(viable FW=41 3; infarcted FW=21 4) in infarcted non-treated animals, and 56 9
25 mm3 (viable FW=37 8; infarcted FW=19 5) in infarcted cytokine-treated
mice.
These values were compared to the expected values of spared and lost
myocardium
at 27 days, given the size of the infarct in the non-treated and cytokine-
treated
animals. From the volume of the LVFW (56 mm3) in SO and infarct size in non-
treated, 62%, and treated, 64%, mice, it was possible to calculate the volume
of
30 myocardium destined to remain (non-treated=21 mm3; treated=20 mm3) and
destined to be lost (non-treated=35 mm3; treated=36 mm3) 27 days after
coronary
occlusion (Fig. 18A). The volume of newly formed myocardium was detected
exclusively in cytokine-treated mice and found to be 14 mm3 (Fig. 18A). Thus,
the

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
61
repair band reduced infarct size from 64% (36 mm3/ 56 nun3=64%) to 39% [(36
min3-14 mm3)/ 56 mm3=39%]. Since the spared portion of the LVFW at 27 days
was 41 and 37 mm3in non-treated and treated mice (see above), the remaining
myocardium, shown in Fig. 18a, underwent 95% (p<0.001) and 85% (p<0.001)
hypertrophy, respectively. Consistently, myocyte cell volume increased 94% and
77% (Fig. 18B).
D. Determination the Total Volume of Formed Myocardium
The volume of regenerating myocardium was determined by measuring in
each of three sections the area occupied by the restored tissue and section
thickness.
The product of these two variables yielded the volume of tissue repair in each
section. Values in the three sections were added and the total volume of
formed
myocardium was obtained. Additionally, the volume of 400 myocytes was
measured in each heart. Sections were stained with desmin and laminin
antibodies
and propidium iodide (PI). Only longitudinally oriented cells with centrally
located
nuclei were included. The length and diameter across the nucleus were
collected in
each myocyte to compute cell volume, assuming a cylindrical shape (Olivetti et
al.,
1991; Beltrami et al., 1994). Myocytes were divided in classes and the number
of
myocytes in each class was calculated from the quotient of total myocyte class
volume and average cell volume (Kajstura et al., 1995; Reiss et al., 1996).
Number
of arteriole and capillary profiles per unit area of myocardium was measured
as
previously done (Olivetti et al., 1991; Beltrami et al., 1994).
Sections were incubated with BrdU or Ki67 antibody. Myocytes (M) were
recognized with a mouse monoclonal anti-cardiac myosin, endothelial cells (EC)
with a rabbit polyclonal anti-factor VIII and smooth muscle cells (SMC) with a
mouse monoclonal anti-a-smooth muscle actin myosin. The fractions of M, EC and
SMC nuclei labeled by BrdU and 1<167 were obtained by confocal microscopy
(Orlic
et al., 2001). Nuclei sampled in 11 cytokine-treated mice; BrdU: M=3,541;
EC=2,604; SMC=1,824. 1<167: M=3,096; EC=2,465; SMC=1,404.
BrdU was injected daily between days 14 to 26 to measure the cumulative
extent of cell proliferation while Ki67 was assayed to determine the number of
cycling cells at sacrifice. Ki67 identifies cells in Gl, S, G2, prophase and
metaphase, decreasing in anaphase and telophase (Orlic et al., 2001). The
percentages of BrdU and Ki67 positive myocytes were 1.6- and 1.4-fold higher
than

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
62
EC, and 2.8- and 2.2-fold higher than SMC, respectively (Fig. 18C, 19). The
forming myocardium occupied 76 11% of the infarct; myocytes constituted
61 12%, new vessels 12 5% and other components 3 2%. The band contained
15x106 regenerating myocytes that were in an active growing phase and had a
wide
size distribution (Figs. 18D-E). EC and SMC growth resulted in the formation
of
5 arterioles and 348 82 capillaries per mm2 of new myocardium. Thick wall
arterioles with several layers of SMC and luminal diameters of 10-30 inn
represented vessels in early differentiation. At times, incomplete perfusion
of the
coronary branches within the repairing myocardium during the fixation
procedure
10 led to arterioles and capillaries containing erythrocytes (Figs. 18F-H).
These results
provided evidence that the new vessels were functionally competent and
connected
with the coronary circulation. Therefore, tissue repair reduced infarct size
and
myocyte growth exceeded angiogenesis; muscle mass replacement was the
prevailing feature of the infarcted heart.
15 E. Determination of Cell Differentiation
Cytoplasmic and nuclear markers were used. Myocyte nuclei: rabbit
polyclonal Csx/Nkx2.5, MEF2, and GATA4 antibodies (Orlic et al., 2001; Lin et
al.,
1997; Kasahara et al., 1998); cytoplasm: mouse monoclonal nestin (Kachinsky et
al.,
1995), rabbit polyclonal desmin (Hermann and Aebi, 1998), cardiac myosin,
mouse
monoclonal a-sarcomeric actin and rabbit polyclonal connexin 43 antibodies
(Orlic
et al., 2001). EC cytoplasm: mouse monoclonal ilk-1, VE-cadherin and factor
VIII
antibodies (Orlic et al., 2001; Yamaguchi et al., 1993; Breier et al., 1996).
SMC
cytoplasm: flk-1 and a-smooth muscle actin antibodies (Orlic et al., 2001;
Couper et
al., 1997). Scar was detected by a mixture of collagen type I and type III
antibodies.
Five cytoplasmic proteins were identified to establish the state of
differentiation of myocytes (Orlic et al., 2001; Kachinsky et al., 1995;
Hermann and
Aebi, 1998): nestin, desmin, a-sarcomeric actin, cardiac myosin and connexin
43.
Nestin was recognized in individual cells scattered across the forming band
(Fig.
20A). With this exception, all other myocytes expressed desmin (Fig. 20B), a-
sarcomeric actin, cardiac myosin and connexin 43 (Fig. 20C). Three
transcription
factors implicated in the activation of the promoter of several cardiac muscle
structural genes were examined (Orlic et al., 2001; Lin et al., 1997; Kasahara
et al.,
1998): Csx/Nkx2.5, GATA-4 and MEF2 (Figs. 21A-C). Single cells positive for

CA 02488346 2011-06-02
63
flk-1 and VE-cadherin (Yamaguchi et al., 1993; Breier et al., 1996), two EC
markers, were present in the repairing tissue (Figs. 20D,E); flk-1 was
detected in
SMC isolated or within the arteriolar wall (Fig. 20F). This tyrosine kinase
receptor
promotes migration of SMC during angiogenesis (Couper et al., 1997).
Therefore,
repair of the infarcted heart involved growth and differentiation of all
cardiac cell
populations resulting in de novo myocardium.
EXAMPLE 3: Migration of Primitive Cardiac Cells in the Adult Mouse Heart
To determine whether a population of primitive cells was present in the adult
ventricular myocardium and whether these cells possessed the ability to
migrate,
three major growth factors were utilized as chemoattractants: hepatocyte
growth
factor (HGF), stem cell factor (SCF) and granulocyte monocyte colony
stimulating
factor (GM-CSF). SCF and GMCSF were selected because they have been shown
to promote translocation of herriatopoietic stem cells. Although HGF induces
migration of hematopoietic stem cells, this growth factor is largely
implicated in
mitosis, differentiation and migration of cardiac cell precursors during early
cardio genesis. On this basis, enzymatically dissociated cells from the mouse
heart
were separated according to their size. Methods for dissociating cardiac cells
from
heart tissue are well-known to those skilled in the art and therefore would
not
involve undue experimentation (CfU.S Patent No. 6,255,292).
A homogenous population of the
dissociated cardiac cells containing small undifferentiated cells, 5-7 pm in
diameter,
with a high nucleus to cytoplasm ratio were subjected to migration assay in
Boyden
microchambers characterized by gelatin-coated filters containing pores, 5 pm
(Boyden et al., 1962, J. Exptl. Med. 115:453-456)
No major differences in the dose-response curve of migrated cells in the
presence of the three growth factors were detected. However, HGF appeared to
mobilize a larger number of cells at a concentration of 100 ng/ml. In
addition, the
cells that showed a chemotactic response to HGF consisted of 15% of c-kit
positive
(c-IdtP s) cells, 50% of multidrug resistance -1 (MDR-1) labeled cells and 30%
of
stem cell antigen-1 (Sca-1) expressing cells. When the mobilized cells were
cultured in 15% fetal bovine serum, they differentiated into myocytes,
endothelial

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
64
cells, smooth muscle cells and fibroblasts. Cardiac myosin positive myocytes
constituted 50% of the preparation, while factor VIII labeled cells included
15%,
alpha-smooth muscle actin stained cells 4%, and vimentin positive factor VIII
negative fibroblasts 20%. The remaining cells were small undifferentiated and
did
not stain with these four antibodies. In conclusion, the mouse heart possesses
primitive cells which are mobilized by growth factors. HGF translocates cells
that in
vitro differentiate into the four cardiac cell lineages.
EXAMPLE 4: Cardiac C-Kit Positive Cells Proliferate In Vitro and Generate New
Myocardium Vivo
To determine whether primitive c-kiPs cells were present in senescent
Fischer 344 rats, dissociated cardiac cells were exposed to magnetic beads
coated
with c-kit receptor antibody (ACK-4-biotin, anti-c-kit mAb). Following
separation,
these small undifferentiated cells were cultured in 10% fetal calf serum.
Cells
attached in a few days and began to proliferate at one week. Confluence was
reached at 7-10 days. Doubling time, established at passage P2 and P4,
required 30
and 40 hours, respectively. Cells grew up to P18 (90th generation) without
reaching
senescence. Replicative capacity was established by Ki67 labeling: at P2, 88
14%
of the cells contained Ki67 protein in nuclei. Additional measurements were
obtained between P1 and P4; 40% of cells expressed alpha-sarcomeric actin or
cardiac myosin, 13% desmin, 3% alpha-smooth muscle actin, 15% factor VIII1 or
CD31, and 18% nestin. Under these in vitro conditions, cells showed no clear
myofibrillar organization with properly aligned sarcomeres and spontaneous
contraction was never observed. Similarly, Ang II, norepinephrine,
isoprotererol,
mechanical stretch and electrical field stimulation failed to initiate
contractile
function. On this basis, it was decided to evaluate whether these cells
pertaining to
the myogenic, smooth muscle cell and endothelial cell lineages had lost
permanently
their biological properties or their role could be reestablished in vivo.
Following
BrdU labeling of cells at P2, infarcted Fischer 344 rats were injected with
these
BrdU positive cells in the damaged region, 3-5 hours after coronary artery
occlusion.
Two weeks later, animals were sacrificed and the characteristics of the
infarcted area
were examined. Myocytes containing parallel arranged myofibrils along their
longitudinal axis were recognized, in combination with BrdU labeling of
nuclei.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
Moreover, vascular structures comprising arterioles and capillary profiles
were
present and were also positive to BrdU. In conclusion, primitive c-kit
positive cells
reside in the senescent heart and maintain the ability to proliferate and
differentiate
into parenchymal cells and coronary vessels when implanted into injured
5 functionally depressed myocardium.
EXAMPLE 5: Cardiac Stem Cells Mediate Myocyte Replication in the Young and
Senescent Rat Heart
The heart is not a post-mitotic organ but contains a subpopulation of
10 myocytes that physiologically undergo cell division to replace dying
cells. Myocyte
multiplication is enhanced during pathologic overloads to expand the muscle
mass
and maintain cardiac performance. However, the origin of these replicating
myocytes remains to be identified. Therefore, primitive cells with
characteristics of
stem/progenitor cells were searched for in the myocardium of of Fischer 344
rats.
15 Young and old animals were studied to determine whether aging had an
impact on
the size population of stem cells and dividing myocytes. The numbers of c-kit
and
MDR1 positive cells in rats at 4 months were 11 3, and 18 6/100 mm2 of tissue,
respectively. Values in rats at 27 months were 35 10, and 42 13/100 mm2. A
number of newly generated small myocytes were identified that were still c-kit
or
20 MDR1 positive. Ki67 protein, which is expressed in nuclei of cycling
cells was
detected in 1.3 0.3% and 4.1 1.5% of myocytes at 4 and 27 months,
respectively.
BrdU localization following 6 or 56 injections included 1.0 0.4% and 4.4 1.2%
at 4
months, and 4.0 1.5% and 16 4% at 27 months. The mitotic index measured in
tissue sections showed that the fraction of myocyte nuclei in mitosis
comprised
25 82 28/106 and 485 98/106 at 4 and 27 months, respectively. These
determinations
were confirmed in dissociated myocytes to obtain a cellular mitotic index. By
this
approach, it was possible to establish that all nuclei of multinucleated
myocytes
were in mitosis simultaneously. This information could not be obtained in
tissue
sections. The collected values showed that 95 31 /106 myocytes were dividing
at 4
30 months and 620 98/106 at 27 months. At both age intervals, the formation
of the
mitotic spindle, contractile ring, disassembly of the nuclear envelope,
karyokinesis
and cytokinesis were documented. In conclusion, primitive undifferentiated
cells
reside in the adult heart and their increase with age is paralleled by an
increase in the

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
66
number of myocytes entering the cell cycle and undergoing karyokinesis and
cytokinesis. This relationship suggests that cardiac stem cells may regulate
the level
and fate of myocyte growth in the aging heart.
EXAMPLE 6: Chimerism of the Human Heart and the Role of stem Cells
The critical role played by resident primitive cells in the remodeling of the
injured heart is well appreciated when organ chimerism, associated with
transplantation of a female heart in a male recipient, is considered. For this
purpose,
8 female hearts implanted in male hosts were analyzed. Translocation of male
cells
to the grafted female heart was identified by FISH for Y chromosome (see
Example
1E). By this approach, the percentages of myocytes, coronary arterioles and
capillary profiles labeled by Y chromosome were 9%, 14% and 7%, respectively.
Concurrently, the numbers of undifferentiated c-kit and multidrug resistance-1
(MDR1) positive cells in the implanted female hearts were measured.
Additionally,
the possibility that these cells contained the Y chromosome was established.
Cardiac transplantation involves the preservation of portions of the atria of
the
recipient on which the donor heart with part of its own atria is attached.
This
surgical procedure is critical for understanding whether the atria from the
host and
donor contained undifferentiated cells that may contribute to the complex
remodeling process of the implanted heart. Quantitatively, the values of c-kit
and,
MDR1 labeled cells were very low in control non-transplanted hearts: 3 c-kit
and 5
MDR1/100 mm2 of left ventricular myocardium. In contrast, the numbers of c-kit
and MDR1 cells in the atria of the recipient were 15 and 42/100 mm2.
Corresponding values in the atria of the donor were 15 and 52/100 mm2 and in
the
ventricle 11 and 21/100 MM2. Transplantation was characterized by a marked
increase in primitive undifferentiated cells in the heart. Stem cells in the
atria of the
host contained Y chromosome, while an average of 55% and 63% of c-kit and
MDR1 cells in the donor's atria and ventricle, respectively, expressed the Y
chromosome. All c-kit and MDR1 positive cells were negative for CD45. These
observations suggest that the translocation of male cells to the implanted
heart has a
major impact on the restructuring of the donor myocardium. In conclusion, stem
cells are widely distributed in the adult heart and because of their
plasticity and

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
67
migration capacity generate myocytes, coronary arterioles and capillary
structures
with high degree of differentiation.
EXAMPLE 7: Identification and Localization of Stem Cells In The Adult Mouse
Heart
Turnover of myocytes occurs in the normal heart, and myocardial damage
leads to activation of myocyte proliferation and vascular growth. These
adaptations
raise the possibility that multipotent primitive cells are present in the
heart and are
implicated in the physiological replacement of dying myocytes and in the
cellular
growth response following injury. On this basis, the presence of
undifferentiated
cells in the normal mouse heart was determined utilizing surface markers
including
c-kit, which is the receptor for stem cell factor, multidrug resistance-1
(MDR1),
which is a P-glycoprotein capable of extruding from the cell dyes, toxic
substances
and drugs, and stem cell antigen-1 (Sca-1), which is involved in cell
signaling and
cell adhesion. Four separate regions consisting of the left and right atria,
and the
base, mid-section and apical portion of the ventricle were analyzed. From the
higher
to the lower value, the number of c-kit positive cells was 2611, 15 5, 10 7
and
6 3/100 mm2 in the atria, and apex, base and mid-section of the ventricle,
respectively. In comparison with the base and mid-section, the larger fraction
of c-
kit positive cells in the atria and apex was statistically significant. The
number of
MDR1 positive cells was higher than those expressing c-kit, but followed a
similar
localization pattern; 43114, 29 16, 14 7 and 12 10/100 mm2in the atria, apex,
base
and mid-section. Again, the values in the atria and apex were greater than in
the
other two areas. Sca-1 labeled cells showed the highest value; 150 36/100 mm2
positive cells were found in the atria. Cells positive for c-kit, MDR1 and Sca-
1 were
negative for CD45, and for myocyte, endothelial cell, smooth muscle cell and
fibroblast cytoplasmic proteins. Additionally, the number of cells positive to
both c-
kit and MDR1 was measured to recognize cells that possessed two stem cell
markers. In the entire heart, 36% of c-kit labeled cells expressed MDR1 and
19% of
MDR1 cells had also c-kit. In conclusion, stem cells are distributed
throughout the
mouse heart, but tend to accumulate in the regions at low stress, such as the
atria and
the apex.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
68
EXAMPLE 8: Repair of Infarcted Myocardium by Resident Cardiac Stem Cells
Migration, Invasion and Expression Assays
The receptor of HGF, c-Met, has been identified on hematopoietic and
hepatic stem cells (126, 90) and, most importantly, on satellite skeletal
muscle cells
(92) and embryonic cardiomyocytes (127). These findings prompted us to
determine
whether c-Met was present in CSCs and its ligand HGF had a biological effect
on
these undifferentiated cells. The hypothesis was made that HGF promotes
migration
and invasion of CSCs in vitro and favors their translocation from storage
areas to
sites of infarcted myocardium in vivo. HGF influences cell migration (128)
through
the expression and activation of matrix metalloproteinase-2 (94, 95). This
enzyme
family may destroy barriers in the extracellular matrix facilitating CSC
movement,
homing and tissue restoration.
IGF-1 is mitogenic, antiapoptotic and is necessary for neural stem cell
multiplication and differentiation (96, 97, 98). If CSCs express IGF-1R, IGF-1
may
impact in a comparable manner on CSCs protecting their viability during
migration
to the damaged myocardium. IGF-1 overexpression is characterized by myocyte
proliferation in the adult mouse heart (65) and this faun of cell growth may
depend
on CSC activation, differentiation and survival.
In the initial part of this study, migration and invasion assays were
conducted
to establish the mobility properties of c-kitPc's and MDR1Pc's cells in the
presence of
the chemotactic HGF.
Cardiac cells were enzymatically dissociated and myocytes were discarded
(124).
Small cells were resuspended in serum-free medium (SFM). Cell migration was
measured by using a modified Boyden chamber that had upper and lower wells
(Neuro Probe, Gaithersburg, MD). The filter for the 48-well plate consisted of
gelatin-coated polycarbonate membrane with pores of 5 i_tm in diameter. The
bottom
well was filled with SFM containing 0.1% BSA and HGF at increasing
concentrations; 50 pi. of small cell suspension were placed in the upper well.
Five
hours later, filters were fixed in 4% paraformaldheyde for 40 minutes and
stained
with PI, and c-kit and MDR1 antibodies. FITC-conjugated anti-IgG was used as a
secondary antibody. Six separate experiments were done at each HGF
concentration.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
69
Forty randomly chosen fields were counted in each well in each assay to
generate a
dose-response curve (fig. 61). The motogenic effects of IGF-1 on small cells
was
excluded by performing migration assays with IGF-1 alone or in combination
with
HGF (data not shown). Invasion assays were done utilizing a chamber with 24-
wells
and 12 cell culture inserts (Chemicon, Temecula, CA). A thin layer of growth
factor-
depleted extracellular matrix was spread on the surface of the inserts.
Conversely,
100 ng/ml of HGF were placed in the lower chamber. Invading cells digested the
coating and clung to the bottom of the polycarbonate membrane. The number of
translocated cells was measured 48 hours later following the same protocol
10_ described in the migration assay. Four separate experiments were done
(fig. 62).
Consistent with the results obtained in the migration assay, IGF-1 had no
effects on
cell invasion (data not shown).
Migration was similar in both cell types and reached its peak at 100 ng/ml
HGF. At 5 hours, the number of c-kitP s and MDR1P s cells transmigrated into
the
lower chamber was 3-fold and 2-fold higher than control cells, respectively.
Larger
HGF concentrations did not improve cell migration (Figures 61 and 62). On this
basis, HGF at 100 ng/ml was also employed to determine the ability of c-kitP s
and
MDR1P s cells to penetrate the synthetic extracellular matrix of the invasion
chamber. In 48 hours, the growth factor increased by 8-fold and 4-fold the
number
of c-ki? s and MDR1P s cells in the lower portion of the chamber (Figures 61
and
62), respectively. IGF-1 had no effect on the mobility of these CSCs at
concentrations varying from 25 to 400 ng/ml. The addition of IGF-1 to HGF did
not
modify the migration and invasion characteristics of c-kitP s and MDR1P s
cells
obtained by HGF alone.
Small, undifferentiated c-MetP s cells were collected with immunomagnetic
beads and the ability of these cells to cleave gelatin was evaluated by
zymography
(Figure 63). Briefly, small cells were isolated from the heart (n=4) and
subsequently
separated by microbeads (Miltenyi, Auburn, CA) coated with c-Met antibody.
Cells
were exposed to HGF, 100 ng/ml, for 30 minutes at 37 C. Cell lysates were run
onto 10% polyacrylamide gels copolymerized with 0.1% gelatin (Invitrogen,
Carlsbad, CA). The gels were incubated in Coomassie blue staining solution
(0.5%)
and areas of gelatinolytic activity were detected as clear bands against a
gray

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
background. This was done to demonstrate whether c-MetP s cells expressed
matrix
metalloproteinases (MMPs) and were capable of digesting the substrate present
in
the gel (94, 95). Positive results were obtained (Figure 63), suggesting that
the
mobility of these primitive cells was due, at least in part, to activation of
MMPs.
Myocardial Infarction in Mice
Myocardial infarction was produced in mice and 5 hours later 4 separate
Female 129 SV-BV mice were used. Following anesthesia (150 mg
ketamine-1 mg acepromazine/kg b.w., i.m.), mice were ventilated, the heart was
exposed and the left coronary artery was ligated (61, 87). Coronary ligation
in
Before discussing the effects of CSCs on organ repair the presence of c-Met
and IGF-1R on cells expressing c-kit and MDR1 was measured in the atria and
left

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
71
ventricle (LV) of control mice. An identical analysis was done in the atria
and
infarcted and non-infarcted LV of mice subjected to coronary artery occlusion.
This
determination was performed 2-3 hours following the administration of growth
factors, which reflected 7-8 hours after coronary occlusion (The objective was
to
document that primitive cells invaded the dead tissue and the surrounding
viable
myocardium and that HGF and IGF-1 were implicated in this process.
c-Met and IGF-1R were detected in c-kiris and MDR1P s cells dispersed in
regions of the normal (n=5), infarcted-treated (n=6) and infarcted-untreated
(n=5)
heart (Fig. 22, A to F). A large fraction of c-kitP s and MDR1P s cells
expressed c-
Met and IGF-1R alone or in combination. Myocardial infarction and the
administration of growth factors did not alter in a consistent manner the
relative
proportion of CSCs with and without c-Met and IGF-1R in the myocardium (Figure
64). Hairpin 1 (apoptosis) and hairpin 2 (necrosis) labeling and K167
expression in
nuclei (cycling cells) were used to establish the viability and activation of
c-kitP s
and MDR1P s cells in the various portions of the damaged and non-damaged
heart,
respectively (Fig. 22, G to L).
CSCs were more numerous in the atria than in the ventricle of control mice.
Acute myocardial infarction and growth factor administration markedly changed
the
number and the distribution of primitive cells in the heart. Viable c-kitP s
and
MDR1P s cells significantly increased in the spared myocardium of the border
zone
and remote tissue as well as in the dead myocardium of the infarcted region.
Importantly, CSCs decreased in the atria (Fig. 22, M and N), suggesting that a
translocation of primitive cells occurred from this site of storage to the
stressed
viable and dead myocardium. A different phenomenon was noted in infarcted-
untreated mice, in which viable CSCs remained higher in the atria than in the
ventricle. In control animals and infarcted-treated mice, apoptosis and
necrosis were
not detected in c-kiPs and MDR1P s cells within the infarct and surrounding
myocardium. Ki67 labeling was identified in nearly 35% and 20% of
undifferentiated cells distributed in the border zone and in the infarct,
respectively
(Figure 65). In infarcted-untreated mice, the majority of c-ki? s and MDR1P 5
cells
in the infarct were apoptotic (Fig. 22, M and N). Necrosis was not seen. An
apoptotic CSC death gradient was observed from the infarct to the distant

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
72
myocardium and atrial tissue. In these mice, only 10-14% of the viable c-
kielps and
MDR1P s cells expressed Ki67 (Figure 65).
Thus, these results support the notion that CSCs express c-Met and IGF-1R
and, thereby, HGF and IGF-1 have a positive impact on the colonization,
proliferation and survival of CSCs in the infarcted heart. On the basis of in
vitro and
in vivo data, HGF appears to have a prevailing role in cell migration and IGF-
1 in
cell division and viability. In infarcted-untreated mice, however, CSCs do not
translocate to the infarcted region and the pre-existing primitive cells die
by
apoptosis. The important question was then whether CSCs located within the
infarct
were capable of differentiating in the various cardiac cell lineages and
reconstitute
dead myocardium. A positive finding would provide a mechanism for cardiac
repair
in infarcted-treated mice and a potential explanation for the absence of
myocardial
regeneration in infarcted-untreated mice.
For anatomical measurements, the heart was arrested in diastole with CdC12,
and the myocardium was perfused with 10% formalin. The LV chamber was filled
with fixative at a pressure equal to the in vivo measured end-diastolic
pressure. The
LV intracavitary axis was determined and the mid-section was used to obtain LV
thickness and chamber diameter. Infarct size was measured by the number of
myocytes lost from the LV inclusive of the interventricular septum (87).
Myocardial infarction at 16 days resulted in a 42% (n=15) and 67% (n=22)
loss of myocytes in the left ventricle and septum of untreated and HGF-IGF-1-
treated mice, respectively (Fig. 23A). In spite of a 60% larger infarct, mice
exposed
to growth factors had a better preservation of cardiac function (Fig. 23B).
HGF-IGF-
1 led to a smaller elevation in LV end-diastolic pressure and a lesser
decrease in
+dP/dt and ¨dP/dt. The difference in infarct size did not influence mortality,
which
was similar in the two groups of mice: 43% in untreated and 40% in treated.
Importantly, 14 of the 22 mice that received growth factors survived with
infarcts
affecting more than 60% of the LV. Seven of these mice had infarcts that
involved
75% to 86% of LV. Untreated mice had infarcts that never exceeded 60% (Fig.
23,
C and D). In contrast to injected mice, a portion of the posterior aspect of
the LV
wall and the entire interventricular septum had to be preserved for untreated
animals
to survive. An infarct larger than 60% is incompatible with life in mice,
rats, dogs

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
73
and any other mammalian species. Irreversible cardiogenic shock and death
supervene in humans with a 46% infarct (99).
From the volume of LV in sham-operated mice and infarct size in untreated
and treated animals it was possible to calculate the volume of myocardium
destined
to remain and destined to be lost 16 days after coronary artery occlusion. The
volume of newly formed myocardium inclusive of myocytes, vascular structures
and
other tissue components was detected exclusively in growth factor-treated mice
and
found to be 8 mm3. Thus, the repair band reduced infarct size from 67% to 57%
(Figures 68 and 69).
The chemotactic and mitogenic properties of HGF-IGF-1 resulted in the
mobilization, proliferation and differentiation of primitive cells in the
infarcted
region of the wall creating new myocardium. In spite of the complexity of this
methodological approach in small animals, the formation of a myocardial band
within the infarct was obtained in 85% of the cases (22 of 26 mice). The band
occupied 65 8% of the damaged area and was located in the mid-portion of the
infarct equally distant from the inner and outer layer of the wall. In very
large
infarcts, the entire thickness of the wall was replaced by developing
myocardium
(Fig. 23, E to H).
Anatomically, the longitudinal axis and the chamber diameter were similar in
the
two groups of infarcted mice indicating that the therapeutic intervention
promoted
positive ventricular remodeling. This notion was consistent with the 60%
larger
infarct size in treated mice. Additionally, the wall thickness-to-chamber
radius ratio
decreased less in treated than in untreated mice. This relationship, in
combination
with the smaller increase in LV end-diastolic pressure in treated mice
significantly
attenuated the increase of diastolic wall stress in this group (Figure 67).
Primitive cells were labeled with monoclonal c-kit and MDR1 antibodies (82,
83). Br
incorporation was detected by BrdU antibody (61, 87). Endothelial cells were
recognized with anti-factor VIII and smooth muscle cells with anti-a-smooth
muscle
actin. For myocyte differentiation, nestin, desmin, cardiac myosin, a-
sarcomeric
actin, N-cadherin and connexin 43 antibodies were utilized. Scar formation in
the
infarct was detected by a mixture of anti-collagen type I and type III (83,
61, 87).).
The composition of the repairing myocardium was evaluated morphometrically.
Antibc

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
74
specific for myocytes, endothelial cells and smooth muscle cells were employed
for
the recognition of parenchymal cells and vessel profiles (61, 87). Moreover,
BrdU
labeling of cells was used as a marker of regenerating tissue over time.
Myocytes
occupied 84 3% of the band, the coronary vasculature 12 3%, and other
structural
components 4 1%. New myocytes varied from 600 to 7,200 [tm3, with an average
volume of 2,200 400 iim3 (Figures 68 and 69). Together, 3.1 1.1 million
myocytes
were formed to compensate for a loss of 2.4 0.8 million cells. This slight
excess in
cell regeneration was at variance with myocyte size. In sham-operated hearts,
myocyte volume, 18,000 3,600 p,m3, was 8.2-fold larger than growing cells.
Importantly, 16% of the muscle mass lost was reconstituted 16 days after
infarction
(lost muscle mass: 18,000 x 2.4 x 106=43 mm3; regenerated muscle mass: 2,200 x
3.1 x 106=7.0 mm3; 7.0:43=16%). The new myocytes were still maturing, but
functionally competent as demonstrated echocardiographically in vivo and
mechanically in vitro.
Echocardiography was performed in conscious mice by using an Acuson
Sequoia 256c equipped with a 13-MHz linear transducer (87). Two-dimensional
images and M-mode tracings were recorded from the parastemal short axis view
at
the level of papillary muscles. Ejection fraction (EF) was derived from LV
cross-
sectional area in 2D short axis view: EF=[(LVDA-LVSA)/LVDA] x 100, where
LVDA and LVSA correspond to LV areas in diastole and systole. For
hemodynamics, mice were anesthetized and a Millar microtip pressure transducer
connected to a chart recorder was advanced into the LV for the evaluation of
pressures and + and - dP/dt in the closed-chest preparation. Echocardiography
performed at day 15 showed that contractile activity was partially restored in
the
regenerating portion of the wall of treated infarcts. Ejection fraction was
also higher
in treated than in untreated mice (Fig. 24, A to E). Thus, structural repair
was
coupled with functional repair.
To confirm that new myocytes reached functional competence and
contributed to the amelioration of ventricular performance, these cells were
enzymatically dissociated from the regenerating myocardium of the infarcted
region
of the wall (129) and their contractile behavior was evaluated in vitro (124,
130).
Myocytes isolated from infarcted treated mice (n=10) by collagenase digestion
were

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
placed in a cell bath (30 0.2 C) containing 1.0 mM Ca2+ and stimulated at 0.5
Hz by
rectangular depolarizing pulses, 3-5 ms in duration in twice diastolic
threshold in
intensity. Parameters were obtained from video images stored in a computer
(124,
130). Developing myocytes were small with myofibrils located at the periphery
of
5 the cell in the subsarcolemmal region. The new myocytes resembled
neonatal cells
actively replicating DNA. They were markedly smaller than the spared
hypertrophied ventricular myocytes (Fig. 25, A and B). In comparison with
surviving old myocytes, growing cells showed a higher peak shortening and
velocity
of shortening, and a lower time to peak shortening (Fig. 25, C to J).
10 The isolated newly generated myocytes were stained by Ki67 to determine
whether these cells were cycling and, therefore, synthesizing DNA. An
identical
protocol was applied to the isolated surviving hypertrophied myocytes of
infarcted-
treated mice. On this basis, the DNA content of each myocyte nucleus in
mononucleated and binucleated cells was evaluated by PI staining and confocal
15 microscopy (see Fig. 25, A and B). Control diploid mouse lymphocytes
were used as
baseline. The objective was to establish if cell fusion occurred in CSCs
before their
commitment to cell lineages. This possibility has recently been suggested by
in vitro
studies (131, 132). Non-cycling new myocytes and enlarged spared myocytes had
only diploid nuclei, excluding that such a phenomenon played a role in cardiac
20 repair (Figure 66).
To establish the level of differentiation of maturing myocytes within the
band, the expression of nestin, desmin, cardiac myosin heavy chain, a-
sarcomeric
actin, N-cadherin and connexin 43 was evaluated. N-cadherin identifies the
fascia
adherens and connexin 43 the gap junctions in the intercalated discs. These
proteins
25 are developmentally regulated. Connexin 43 is also critical for
electrical coupling
and synchrony of contraction of myocytes. These 6 proteins were detected in
essentially all newly formed myocytes (Fig. 26, A to N). The percentage of
myocytes labeled by BrdU was 84 9%, indicating that cell proliferation was
ongoing in the regenerating tissue. Cardiac repair included the formation of
30 capillaries and arterioles (Fig. 27, A to D). The presence of red blood
cells within
the lumen indicated that the vessels were connected with the coronary
circulation.
This phase of myocardial restoration, however, was characterized by a
prevailing

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
76
growth of resistance arterioles than capillary structures. There were 59 29
arterioles
and 137 80 capillaries per mm2 of new myocardium.
The current findings indicate that resident CSCs can be mobilized from their
region of storage to colonize the infarcted myocardium where they
differentiate into
cardiac cell lineages resulting in tissue regeneration. The intervention
utilized here
was capable of salvaging animals with infarct size normally incompatible with
life
in mammals.
EXAMPLE 9: Cardiac Stem Cells Differentiate In Vitro Acquiring Functional
Competence In Vivo
A. Collection and Cloning of Cells
Cardiac cells were isolated from female Fischer rats at 20-25 months of age
(111, 112). Intact cells were separated and myocytes were discarded. Small
cells
were resuspended and aggregates removed with a strainer. Cells were incubated
with
a rabbit c-kit antibody (H-300, Santa Cruz) which recognizes the N-terminal
epitope
localized at the external aspect of the membrane (121). Cells were exposed to
magnetic beads coated with anti-rabbit IgG (Dynal) and c-kitP s cells were
collected
with a magnet (n=13). For FACS (n=4), cells were stained with r-phycoerythrin-
conjugated rat monoclonal anti-c-kit (Pharmingen). With both methods, c-ki? s
cells
varied from 6-9% of the small cell population.
c-ki? s cells scored negative for myocyte (a-sarcomeric actin, cardiac
myosin, desmin, a-cardiac actinin, connexin 43), endothelial cell (EC; factor
VIII,
CD31, vimentin), smooth muscle cell (SMC; a-smooth muscle actin, desmin) and
fibroblast (F; vimentin) cytoplasmic proteins. Nuclear markers of myocyte
lineage
(Nkx2.5, MEF2, GATA-4) were detected in 7-10% and cytoplasmic proteins in 1-
2% of the cells. c-kit' s cells did not express skeletal muscle transcription
factors
(MyoD, myogenin, Myf5) or markers of the myeloid, lymphoid and erythroid cell
lineages (CD45, CD45RO, CD8, TER-119), indicating the cells were Lin-c-kiPs
cells.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
77
c-kitP s cells were plated at 1-2x104 cells/ml NSCM utilized for selection and
growth of neural stem cells (122). This was composed by Dulbecco's MEM and
Ham's F12 (ratio 1:1), bFGF, long/ml, EGF, 2Ong/ml, HEPES, 5mM, insulin-
transferrin-selenite. c-ki? s cells attached in two weeks and began to
proliferate
(Fig. 28a,b). NSCM was then substituted with differentiating medium (DM) and
confluence was reached in 7-10 days. Cells were passaged by trypsinization.
Cycling cells, as determined by Ki67 expression, varied from 74 12% to 84 8%
at
passages (P) P1-P5 (n=5 at each P). Doubling time at P2 and P4 averaged 41
hours.
Cells continued to divide up to P23 without reaching growth arrest and
senescence,
at which time cells were frozen. Cardiac lineages were identified from PO to
P23.
At PO (n=7), P3 (n=10), P10 (n=13) and P23 (77=13), myocytes were 29-40%, EC
20-26%, SMC 18-23% and F 9-16%. Aliquots of P23 grown after 6 months in liquid
nitrogen expressed the same phenotypes as the parental cells.
At PO and P1 when grown in DM, 50% of the cells exhibited Nkx2.5, 60%
MEF2, 30% GATA-4 and 55% GATA-5 (Fig. 28c-f). Conversely, skeletal muscle
(MyoD, myogenin, Myf5), blood cell (CD45, CD45RO, CD8, TER-119) and neural
(MAP lb, neurofilament 200, GFAP) markers were not identified.
For cloning, cells were seeded at 10-50 cells/ml NSCM (Fig. 28g) (109,
110). After one week, colonies derived from a single cell were recognized
(Fig.
28h); fibronectin, procollagen type I and vimentin were absent excluding the
fibroblast lineage. Individual colonies were detached with cloning cylinders
and
plated. Multiple clones developed and one clone in each preparation was chosen
for
characterization. MEM containing 10% FCS and 10-8 M dexamethasone was
employed to induce differentiation (DM). For subcloning, cells from multiple
clones
were plated at 10-50 cells/ml NSCM. Single subclones were isolated and plated
in
DM. At each subcloning step, an aliquot of cells was grown in suspension to
develop clonal spheres.
Each clone contained groups of 2-3 Lin-c-ki? s cells (Fig. 29a), although the
majority of these cells (-20-50) were dispersed among c-kitNEG cells. Some
cells
were Ki67 positive and occasionally in mitosis (Fig. 29b-d). Myocytes
expressing
cardiac myosin and a-sarcomeric actin, EC expressing factor VIII, CD31 and

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
78
vimentin, SMC expressing a-smooth muscle actin and F expressing vimentin alone
were identified in each clone (Fig. 29e-h). Aggregates of small cells
containing
nestin were also present (Supplementary Information). Thus, Lin-c-kitP s cells
isolated from the myocardium possessed the properties expected for stem cells.
They
were clonogenic, self-renewing and multipotent and gave origin to the main
cardiac
cell types. Subclonal analysis of several primary clones confirmed the
stability of the
phenotype of the primary clones: clonogenicity, self-renewal and
multipotentiality.
The phenotype of most subclones was indistinguishable from that of the primary
clones. However, in two of eight subclones, only myocytes were obtained in one
case and exclusively EC were identified in the other.
Clonogenic cells, grown in suspension in Corning untreated dishes generated
spherical clones (Fig. 30a). This anchorage independent growth is typical of
stem
14,15 .../ci?OS and c_kitNEG
cells '4"5. Spheroids consisted of clusters of c cells
and large
amounts of nestin (Fig. 30b-d). Similarly to other stem cells14'15, following
plating in
DM, spheroids readily attached, and cells migrated out of the spheres and
differentiated (Fig. 30e-h).
Cells were fixed in 4% paraformaldehyde and undifferentiated cells were
labeled with c-kit antibody. Markers for myocytes included Nkx2.5, MEF2, GATA-
4, GATA-5, nestin, a-sarcomeric actin, a-cardiac actinin, desmin and cardiac
myosin heavy chain. Markers for SMC comprised a-smooth muscle actin and
desmin, for EC factor VIII, CD31 and vimentin, and for F vimentin in the
absence of
factor VIII, fibronectin and procollagen type I. MyoD, myogenin and Myf5 were
utilized as markers of skeletal muscle cells. CD45, CD45RO, CD8 and TER-119
were employed to exclude hematopoietic cell lineages. MAP lb, neurofilament
200
and GFAP were used to recognize neural cell lineages. BrdU and Ki67 were
employed to identify cycling cells (61, 87). Nuclei were stained by PI.
Myocytes and SMC failed to contract in vitro. Angiotensin II, isoproterenol,
norepinephrine and electrical stimulation did not promote contraction. EC did
not
express markers of full differentiation such as eNOS.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
79
B. Myocardial infarction and cell implantation
BrdU labeled cells (P2; positive cells=88 6%) were implanted. Myocardial
infarction was produced in female Fischer rats at 2 months of age (111). Five
hours
later, 22 rats were injected with 2x105 cells in two opposite regions
bordering the
infarct; 12 rats were sacrificed at 10 days and 10 rats at 20 days. At each
interval, 8-
9 infarcted and 10 sham-operated rats were injected with saline and 5 with Lin-
c-
kitNEG cells and used as controls. Under ketamine anesthesia, echocardiography
was
performed at 9 and 19 days, only in rats killed at 20 days. From M-mode
tracings,
LV end-diastolic diameter and wall thickness were obtained. Ejection fraction
was
computed (87). At 10 and 20 days, animals were anesthetized and LV pressures
and
+ and - dP/dt were evaluated in the closed-chest preparation (111). Mortality
was
lower but not statistically significant in treated than in untreated rats at
10 and 20
days after surgery, averaging 35% in all groups combined. Protocols were
approved
by the institutional review board.
C. Anatomic and Functional Results
Hearts were arrested in diastole and fixed with formalin. Infarct size was
,
determined by the fraction of myocytes lost from the left ventricle (87), 53
7% and
49 10% (NS) in treated and untreated rats at 10 days, and 70 9% and 55 10%
(P<0.001) in treated and untreated rats at 20 days, respectively. The volume
of 400
new myocytes was measured in each heart. Sections were stained with desmin and
laminin and PI. In longitudinally oriented myocytes with centrally located
nuclei,
cell length and diameter across the nucleus were collected to compute cell
volume
(87).
Sections were incubated with BrdU and Ki67 antibodies. A band of
regenerating myocardium was identified in 9 of 12 treated infarcts at 10 days,
and in
all 10 treated infarcts at 20 days. At 10 days, the band was thin and
discontinuous
and, at 20 days, was thicker and present throughout the infarcted area (Figure
31a-
c). Myocytes (M), EC, SMC and F were identified by cardiac myosin, factor
VIII,

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
a-smooth muscle actin and vimentin in the absence of factor VIII,
respectively.
Myocytes were also identified by cardiac myosin antibody and propidium iodide
(PI). At 10 and 20 days, 30 and 48 rnm3 of new myocardium were measured,
respectively. Tissue regeneration reduced infarct size from 53 7% to 40 5%
5 (P<0.001) at 10 days, and from 70 9% to 48 7% (P<0.001) at 20 days
Cells labeled by BrdU and Ki67 were identified by confocal microscopy
(103, 105). The number of nuclei sampled for BrdU labeling were: M=5,229;
EC=3,572; SMC=4,010; F=5,529. Corresponding values for Ki67 were: M=9,290;
EC=9,103; SMC=8,392. Myocyte differentiation was established with cardiac
10 myosin, a-sarcomeric actin, a-cardiac actinin, N-caclherin and connexin
43.
Collagen was detected by collagen type I and type III antibodies.
Since implanted cells were labeled by BrdU, the origin of the cells in the
developing myocardium was identified by this marker. Myocytes, arterioles
(Fig.
3 if-n) and capillary profiles were detected. At 10 days, the proportion of
myocytes,
15 capillaries and arterioles was lower, and collagen was higher than at 20
days. Cell
growth evaluated by Ki67 was greater at 10 days decreasing at 20 days
(Supplementary Information).
Cardiac myosin, a-sarcomeric actin, a-cardiac actinin, N-cadherin and
connexin 43 were detected in myocytes (Fig. 3 lm-t; Supplementary
Information). At
20 10 days, myocytes were small, sarcomeres were rarely detectable and N-
cadherin
and connexin 43 were mostly located in the cytoplasm (Fig. 31nz,n,q,r).
Myocyte
volume averaged 1,5001.un3 and 13.9x106 myocytes were formed. At 20 days,
myocytes were closely packed and myofibrils were more abundant; N-cadherin and
connexin 43 defined the fascia adherens and nexuses in intercalated discs
(Fig.
25 31o,p,s,t). Myocyte volume averaged 3,400 inn3 and 13x106myocytes were
present
Myocyte apoptosis was measured by in situ ligation of hairpin
oligonucleotide probe with single base overhang. The number of nuclei sampled
for
apoptosis was 30,464 at 10 days and 12,760 at 20 days. The preservation of
myocyte
number from 10 to 20 days was consistent with a decrease in Ki67 labeling and
an

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
81
increase in apoptosis (0.33 0.23% to 0.85 0.31%, P<0.001). Thus, myocyte
proliferation prevailed early and myocyte hypertrophy later. From 10-20 days,
the
number of vessels nearly doubled.
Procedures for determining mechanical properties of the new myocytes have
been previously described30. Myocytes isolated from infarcted treated rats
(n=4)
were placed in a cell bath (30 0.2 C) containing 1.0 mM Ca2+ and stimulated at
0.5
Hz by rectangular depolarizing pulses, 3-5 ms in duration in twice diastolic
threshold in intensity. Mechanical parameters were obtained from video images
stored in a computer. The mechanical behavior of myocytes isolated from the
infarcted and non-infarcted regions of treated hearts was measured at 20 days
(Fig.
32a-e). New cells were calcium tolerant and responded to stimulation. However,
in
comparison with spared myocytes, maturing cells showed a decreased peak
shortening and velocity of shortening; time to peak shortening and time to 50%
re-
lengthening were similar in the two groups of cells (Fig. 33a-/). Developing
myocytes had myofibrils mostly distributed at the periphery; sarcomere
striation was
apparent (Fig. 32a-e).
Cell implantation reduced infarct size and cavitary dilation, and increased
wall thickness and ejection fraction. Contraction reappeared in the infarcted
ventricular wall and end-diastolic pressure, developed pressure and + and ¨
dP/dt
improved at 20 days. Diastolic stress was 52% lower in treated rats
(Supplementary
Information). Thus, structural and functional modifications promoted by
cardiac
repair decreased diastolic load and ameliorated ventricular performance. This
beneficial effect occurred in spite of the fact that infarct size was similar
in the two
groups of rats.
Colonization, replication, differentiation of the transplanted cells and
tissue
regeneration required c-kites cells and damaged myocardium. c-kiPs cells
injected
in sham-operated rats grafted poorly and did not differentiate. Injection of c-
kitNEG
cells in the border of infarcts had no effect on cardiac repair.
The multipotent phenotype of the Lin-c-ki?Gs cell reported here is in
apparent contrast with cardiac cell lineage determinations in chicken (113),
zebrafish
(114) and mammals (115) concluding that myocytes, SMC, and EC each originates
from a separate lineage. However, not all studies are in agreement (116).
Because

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
82
these experiments (113, 114, 115, 116) did not address the developmental
potential
of any of the cells marked, as has been done here, the different outcomes
likely
represent another example of the difference between normal developmental fate
and
developmental potential. Additionally, the plasticity of human embryonic stem
cells
(117), progenitor endothelial cells (101) and clonogenic cells (52) as means
to repair
damaged myocardium has recently been documented (101,52).
EXAMPLE 10: Mobilization of Cardiac Stem Cells (CSC) by Growth Factors
Promotes Repair of Infarcted Myocardium Improving Regional and Global Cardiac
Function in Conscious Dogs
The methods of the previous non-limiting examples were used with
exceptions as described below.
Myocardial regeneration after infarction in rodents by stem cell homing and
differentiation has left unanswered the question whether a similar type of
cardiac
repair would occur in large mammals. Moreover, whether new myocardium can
affect the functional abnormality of infarcted segments restoring contraction
is not
known. For this purpose, dogs were chronically instrumented for measurements
of
hemodynamics and regional wall function. Stroke volume and EF were also
determined. Myocardial infarction was induced by inflating a hydraulic
occluder
around the left anterior descending coronary artery. Four hours later, HGF and
IGF-
1 were injected in the border zone to mobilize and activate stem cells; dogs
were
then monitored up to 30 days. Growth factors induced chronic cardiac repair
reversing bulging of the infarct: segment shortening increased from -2.0 0.7%
to
+5.5 2.2%, stroke work from -18 11 to +53 10 mm x mmHg, stroke volume from
22 2 to 45 4 ml and ejection fraction from 39 3 to 64 4%. In treated dogs at 8
hours after infarction, the number of primitive cells increased from 240 40 c-
kit
positive cells at baseline to 1700 400 (remote myocardium), 4400 1200 (border
zone) and 3100 900 c-kit positive cells/100 mm2 (infarcted area). Ki67
labeling was
detected in 48%, 46% and 26% of c-kit positive cells in the remote, border and
infarcted myocardium, respectively. Thus, high levels of these cells were
replicating.
These effects were essentially absent in infarcted untreated dogs. Acute
experiments
were complemented with the quantitative analysis of the infarcted myocardium

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
83
defined by the implanted crystals 10-30 days after coronary occlusion. Changes
from paradoxical movement to regular contraction in the new myocardium were
characterized by the production of myocytes, varying in size from 400 to
16,000
with a mean volume of 2,000 640 p,m3. Resistance vessels with BrdU-labeled
endothelial and smooth muscle cells were 87 48 per mm2 of tissue. Capillaries
were
2-3-fold higher than arterioles. Together, 16 9% of the infarct was replaced
by
healthy myocardium. Thus, canine resident primitive cells can be mobilized
from the
site of storage to reach dead myocardium. Stem cell activation and
differentiation
promotes repair of the infarcted heart improving local wall motion and
systemic
hemodynamics.
EXAMPLE 11: Mobilization of Resident Cardiac Stem Cells Constitutes an
Important Additional Treatment to Angiotensin II Blockade in the Infarcted
Heart
The methods of the previous non-limiting examples were used with
exceptions as described below.
Two of the major complicating factors of myocardial infarction (MI) are the
loss of muscle mass and cavitary dilation, which both contribute to negative
left
ventricular (LV) remodeling and to the depression in cardiac performance. In
an
attempt to interfere with these deleterious effects of MI, resident cardiac
stem cells
(CSC) were mobilized and activated to promote tissue regeneration, and the ATi
receptor blocker losartan (Los) was administered, 20 mg/kg body weight/day, to
attenuate cellular hypertrophy, and, thereby, the expansion in chamber volume.
On
this basis, MI was produced in mice and the animals were subdivided in four
groups:
1. Sham-operated (SO); 2. MI only; 3. MI-Los; 4. MI-Los-CSC. One month after
MI, animals were sacrificed, and LV function, infarct dimension and cardiac
remodeling were evaluated. Myocardial regeneration was also measured in mice
treated with CSC. Infarct size, based on the number of myocytes lost by the LV
was
47% in MI, 51% MI-Los and 53% MI-Los-CSC. In comparison with MI and MI-
Los, MI treated with Los and CSC resulted in a more favorable outcome of the
damaged heart in terms of chamber diameter: -17% vs MI and -12% vs MI-Los;
longitudinal axis: -26% (p<0.001) vs MI and -8% (p<0.02) vs MI-Los; and
chamber
volume: -40% (p<0.01) vs MI and -35% (p<0.04) vs MI-Los. The LV-mass-to-

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
84
chamber volume ratio was 47% (p<0.01) and 56% (p<0.01) higher in MI-Los-CSC
than in MI and MI-Los, respectively. Tissue repair in MI-Los-CSC was made of
10
x 106 new myocytes of 9001.tm3. Moreover, there were 70 arterioles and 200
capillaries per mm2 of myocardium in this group of mice. The production of 9
mm3
of new myocardium reduced MI size by 22% from 53% to 41% of LV.
Echocardiographically, contractile function reappeared in the infarcted region
of the
wall of mice with MI-Los-CSC. Hemodynamically, MI-Los-CSC mice had a lower
LVEDP, and higher + and -dP/dt. In conclusion, the positive impact of losartan
on
ventricular remodeling is enhanced by the process of cardiac repair mediated
by
translocation of CSC to the infarcted area. Mobilized CSC reduce infarct size
and
ventricular dilation and, thereby, ameliorate further the contractile behavior
of the
infarcted heart.
EXAMPLE 12: Hepatocyte Growth Factor (HGF) Induces the Translocation of c-
met to the Nucleus Activating the Expression of GATA-4 and Cardiac Stem Cell
(CSC) Differentiation
The methods of the previous non-limiting examples were used with
exceptions as described below.
In preliminary studies we were able to document that CSCs positive for c-kit
or MDR-1 expressed the surface receptor c-met. c-met is the receptor of HGF
and
ligand binding promoted cell motility via the synthesis of matrix
metalloproteinases.
However, it was unknown whether c-met activation had additional effects on
CSCs
biology and function. For this purpose, we tested whether c-met on CSCs
exposed to
50 ng/ml of HGF in NSCM responded to the growth factor by internalization and
translocation within the cell. Surprisingly, a localization of c-met in the
nucleus was
detected by confocal microscopy in these stimulated cells which maintained
primitive characteristics. This unusual impact of HGF on c-met raised the
possibility
that the mobilized receptor could interact with other nuclear proteins
participating in
cell growth and differentiation of CSCs. Because of the critical role of the
cardiac
specific transcription factor GATA-4 in the commitment of cell lineage. By
immunoprecipitation and Western blot, a protein complex made by c-met and
GATA-4 was identified. A time-dependent analysis following a single HGF
stimulation showed a progressive increase in c-met-GATA-4 complex from 15

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
minutes to 3 days. Time was also coupled with differentiation of primitive
cells into
myocytes and other cardiac cells. To establish a molecular interaction at the
DNA
level between GATA-4 and c-met, a gel retardation assay was performed on
nuclear
extracts isolated from cells stimulated with HGF for 1 hour. A shifted band
was
5 obtained utilizing a probe containing the GATA sequence. However, the
addition of
GATA-4 antibody resulted in a sup ershifted band. Conversely, the inclusion of
c-
met antibody attenuated the optical density of the GATA band. Since a GATA
sequence upstream to the TATA box was identified in the c-met promoter, a
second
mobility shift assay was performed. In this case, nuclear extracts from HGF
10 stimulated cells resulted in a shifted band which was diminished by c-
met antibody.
In contrast, GATA-4 antibody induced a supershifted band. Thus, HGF-mediated
translocation of c-met at the level of the nucleus may confer to c-met a
function of
transcription factor and future studies will demonstrate whether this DNA
binding
enhances the expression of GATA-4 leading to the differentiation of immature
15 cardiac cells.
* * *
Having thus described in detail preferred embodiments of the present
invention, it is to be understood that the invention defined by the appended
claims is
20 not to be limited by particular details set forth in the above
description as many
apparent variations thereof are possible without departing from the spirit or
scope
thereof.
25 REFERENCES
1. American Heart Association. 2001 Heart and Stroke Statistical Update.
Dallas, Texas: American Heart Association, 2000.
2. Bautz, F. et al., "Expression and secretion of vascular endothelial
growth
30 factor-A by cytokine stimulated hematopoietic progenitor cells. Possible
role
in the hematopoietic microenvironment." Exp Hematol 2000 June; 28(6):700-
6.
3. Beardsle, M. A. et al., "Rapid turnover of connexin43 in the adult rat
heart."
Circ. Res. (1998) 83, 629-635.
35 4. Beltrami, C.A. et al., "Structural basis of end-stage failure in
ischemic
cardiomyopathy in humans." Circulation (1994) 89, 151-163.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
86
5. Bianco, P. et al. "Bone marrow stromal stem cells: nature, biology, and
potential applications." Stem Cells (2001) 19:180-192.
6. Blume et al., "A review of autologous hematopoetic cell
transplantation."
Biology of Blood & Marrow Transplantation, (2000) 6: 1-12.
7. Bodine, D.M. et al., "Efficient retrovirus transduction of mouse
pluripotent
hematopoietic stem cells mobilized into the peripheral blood by treatment with
granulocyte colony-stimulating factor and stem cell factor." Blood (1994) 84,
1482-1491.
8. Breier, G. et al., "Molecular cloning and expression of murine vascular
endothelial-cadherin in early stage development of cardiovascular system."
Blood (1996) 87, 630-641.
9. Brugger et al., "Ex vivo manipulation of hematopoetic stem and
progenitor
cells. Seminars in Hematology." (2000), 37 (1): 42-49.
10. Caceres-Cortes, J.R. et al., "Steel factor sustains SCL expressionand
the
survival of purified CD34+ bone marrow cells in the absence of detectable cell
differentiation." Stem Cells (2001) January;19(1):59-70.
11. Caplan A.I. and Haynesworth S.E., "Method for enhancing the
implantation
and differentiation of marrow-derived mesenchymal cells." Filed November
16, 1990. U.S. Patent No. 5,197,985
12. Chin et al., "Cellular Cardiomyoplasty: Mycardial Regeneration With
Satellite
Cell Implantation." Ann. Thorac. Surg. (1995) 60: 12-18.
13. Clutterbuck, R.D. et al., "G-CSF mobilization of haemopoietic cell
populations in SOD mice engrafted with human leukaemia." Bone Marrow
Transplant (1997) August; 20(4):325-32.
14. Coles, J.G. et al., "Inhibition of Human Xenogenic or Allogenic Antibodies
to
Reduce Xenograft or Allograft Rejection in Human Recipients". Patent No.
WO 95/34581A1, published December 21, 1995.
15. Couper, L.L. et al., "Vascular endothelial growth factor increases the
mitogenic response to fibroblast growth factor-2 in vascular smooth muscle
cells in vivo via expression of fms-like tyrosine kinase-1." (1997) Circ. Res.
81, 932-939.
16. Dinsmore, J. "Procine Cardiomyocytes and Their Use in Treatment of
Insufficient Cardiac Function". Patent No. WO 96/38544, published
December 5, 1996.
17. Durocher, D. et al., "The cardiac transciption factors 1\1)(2-5 and GATA-4
are
mutual cofactors." EMBO J. 16, 5687-5696 (1997).
18. Field L.J. "Non-human mammal having a graft and methods of delivering
protein to myocardial tissue." Filed November 16, 1992. U.S. Patent No. 5,
602,301.
19. Field L.J. "Myocardial grafts and cellular compositions." Filed June 7,
1995.
U.S. Patent No. 5,733,727.
20. Field, L.J. "Myocardial Grafts and Cellular Compositions Useful for
Same."
Patent No. WO 95/14079A1, published May 26, 1995.
21. Fielding et al., "Autologous bone marrow transplantation." Curr. Opin.
Hematology, 1994, 1: 412-417.
22. Gillis S. "Method for improving autologous transplantation." Filed
September
26, 1991. U.S. Patent No. 5,199,942
23. Gussoni et al., "Normal dystrophin transcripts detected in Duchenne
muscular
dystrophy patients after myoblast transplantation." Nature 356:435-438 (1992).

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
87
24. Hermann, H. and Aebi, U. "In Subcellular Biochemistry: Intermediate
Filaments." Vol. 31 (ed. Herrmann, H. & Harris, E.) 319-362 (Plenum Press,
New York, 1998).
25. Huang H.M. et al., "Optimal proliferation of a hematopoietic progenitor
cell
line requires either costimulation with stem cell factor or increase of
receptor
expression that can be replaced by overexpression of Bc1-2. Blood." 1999 Apr
15;93(8):2569-77.
26. Ikuta, K. et al., "Mouse hematopoietic stem cells and the interaction
of c-kit
receptor and steel factor." International Journal of Cell Cloning 1991; 9:451-
460.
27. Janowska-Wieczorek, A. et al., "Autocrine/paracrine mechanisms in human
hematopoiesis." Stem Cells 2001; 19:99-107.
28. Jo, D.Y. et al., "Chemotaxis of primitive hematopoietic cells in
response to
stromal cell-derived factor-1." The Journal of Clinical Investigation 2000
January; 105(4101-111.
29. Kachinsky, A.M. et al., "Intermediate filaments in cardiac myogenesis:
nestin
in the developing mouse heart." (1995) J. Histochem. Cytochem. 43, 843-847.
30. Kanj et al., "Myocardial ischemia associated with high-dose carmustine
infusion." Cancer, 1991, 68 (9): 1910-1912.
31. Kajstura, J. et al., "The cellular basis of pacing-induced dilated
cardiomyopathy. Myocyte cell loss and myocyte cellular reactive
hypertrophy." (1995) Circulation 92, 2306-2317.
32. Kasahara, H. et al., "Cardiac and extracardiac expression of
Csx/Nkx2.5
homeodomain protein." (1998) Circ. Res. 82, 936-946.
33. Kedes, L.H. et al., "Compositions and Methods for Transduction of Cells."
Patent No. WO 95/12979A1, published May 18, 1995.
34. Keil F. et al., "Effect of interleukin-3, stem cell factor and
granulocyte-
macrophage colony-stimulating factor on committed stem cells, long-term
culture initiating cells and bone marrow stroma in a one-step long-term bone
marrow culture." Ann Hematol. 2000 May;79(5):243-8.
35. Kempermann, G. et al., "Activity-dependent regulation of neuronal
plasticity
and self repair." Frog Brain Res 2000; 127:35-48.
36. Kim, C.H. and Broxmeyer H.E., "In vitro behavior of hematopoietic
progenitor cells under the influence of chemoattractants: stromal cell-derived
factor-1, steel factor, and the bone marrow environment." Blood 1998 Jan 1;
91(4100-10.
37. Kocher, A.A. et al., "Neovascularization of ischemic myocardium by
human
bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis reduces
remodeling and improves cardiac function." Nature Medicine 2001 April;
7(4):430-436.
38. Koh et al., "Differentiation and long-term survival of C2C12 myoblast
grafts
in heart." Journal of Clinical Investigation 92:1548-1554 (1993).
39. Krause, D.S. et al., "Multi-organ, multi-lineage engraftment by a
single bone
marrow-derived stem cell." Cell (2001) May;105(3)369-370.
40. Kronenwett, R. et al., "The role of cytokines and adhesion molecules for
mobilization of peripheral blood stem cells." Stern Cells 2000; 18:320-330.
41. LaIuppa, J.A. et al., "Evaluation of cytokines for expansion of the
megakaryocyte and ranulocyte lineages." Stern Cells (1997) May:15(3):198-
206.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
88
42. Leor et al., "Transplantation of Fetal Myocardial Tissue Into the
Infarcted
Myocardium of Rat, A Potential Method for Repair of Infarcted
Myocardium?" Circulation 94:(Supplement II) 11-332 - 11-336 (1996).
43. Li et al., "Method of Culturing Cardiomyocytes from Human Pediatric
Ventricular Myocardium." (1992) 1 Tiss. Cult. Meth.; 93-100.
44. Li, Q. et al. "Overexpression of insulin-like growth factor-1 in mice
protects
from myocyte death after infarction, attenuating ventricular dilation, wall
stress, and cardiac hypertrophy." J Clin Invest. 100, 1991-1999 (1997).
45. Li, B et al., "Insulin-like growth factor-1 attenuates the detrimental
impact of
nonocclusive coronary artery constriction on the heart." (1999) Circ. Res. 84,
1007-1019.
46. Li et al., J. Mol. Cell. Cardiol., 26:A162 (1994).
47. Li et al., Cardiovascular Res. 32:362-373 (1996).
48. Li et al., "In Vivo Survival and Function of Transplanted Rat
Cardiomyocytes"
Circulation Research 78:283-288 (1996).
49. Li et al., "Cardiomyocyte Transplantation Improves Heart Function"
(1996)
The Society of Thoracic Surgeons; 62: 654-661.
50. Li et al., "Human Pediatric and Adult Ventricular Cardiomyocytes in
Culture:
Assessment of Phenotypic Changes with Passaging" Feb. 20, 1996
Cardiovascular Research; 1-12.
51. Lin, Q. et at., "Control of mouse cardiac morphogenesis and myogenesis
by
transcription factor MEF2C." (1997) Science 276, 1404-1407.
52. Malouf, N.N. et al., "Adult derived stem cells from the liver become
myocytes
in the heart in vivo." Am J Pathology 2001 June; 158(6)1929-35.
54. Morin, S. et al., "GATA-dependent recruitment of MEF2 proteins to
target
promoters." (2000) EMBO J. 19, 2046-2055.
55. Murray et al., "Skeletal Myobalst Transplantation for Repair of
Myocardial
Necrosis" J. Clin. Invest. 98:2512-2523 (1996).
57. National Institutes of Health. "Stem Cells : A Primer." National
Insitutes of
Health: May 2000.
59. Olivetti, G. et al., "Cellular basis of chronic ventricular
remodeling after
myocardial infarction in rats." (1991) Circ. Res. 68(3), 856-869.
61. Orlic, D. et al., "Bone marrow cells regenerate infarcted myocardium."
(2001)
Nature 410, 701-705.
62. Patchen, ML et al. "Mobilization of peripheral blood progenitor cells
by
Betafectin PGG-glucan alone and in combination with granulocyte colony-
45 stimulating factor." Stem Cells (1998) May; 16(3):208-217.
63. Pfeffer, M. A. and Braunwald, E. "Ventricular remodeling after
myocardial
infarction." Circulation 81, 1161-1172 (1990).
64. Pollick, C. et al., "Echocardiographic and cardiac Doppler assessment
of
mice." (1995) 1 Am. Soc. Echocardiogr. 8, 602-610 (1995).

CA 02488346 2004-12-03
WO 03/103611 PCT/US03/07731
89
65. Reiss, K. et al., "Overexpression of insulin-like growth factor-1 in
the heart is
coupled with myocyte proliferation in transgenic mice." (1996) Proc. NatL
Acad. Sci. USA 93(16), 8630-8635.
66. Roberts M.M., et al., "Prolonged release and c-kit expression of
haemopoietic
precursor cells mobilized by stem cell factor and granulocyte colony
stimulating factor." Br J Haematol. 1999 Mar;104(4):778-84.
67. Rosenthal, IN. and Tsao, L. "Helping the heart to heal with stem
cells."
Nature Medicine 2001 April; 7(4):412-413.
68. Scholzen, T., and Gerdes, J. "The ki-67 protein: from the known and the
unknown." J. Cell. PhysioL 182, 311-322 (2000).
69. Shimomura T., et al., "Thrombopoietin stimulates murine lineage
negative,
Sca-1+, C-Kit+, CD34- cells: comparative study with stem cell factor or
interleukin-3." Int J Hematol. (2000) Jan;71(1):33-9.
70. Soonpaa et al. "Formation of nascent intercalated disks between grafted
fetal
cardiomyocytes and host myocardium." (1994) Science 264(5155):98-101.
71. Silver J. et al., "Methods of reducing glial scar formation and
promoting axon
and blood vessel growth and/or regeneration through the use of activated
immature astrocytes." Filed October 27, 1989. U.S. Patent No. 5,202,120.
72. Simnett et al. "Autologous stem cell translantation for malignancy: a
systemic
review of the literature." Clin. Lab Haem. 2000, 22:61-72.
73. Smith D.A. et al., "Method for inducing human myocardial cell
proliferation."
Filed April 4, 1995. U.S. Patent No. 5,580,779
74. Smith D.A. and Townsend LE. "Method of isolation, culture and
proliferation of human atrial myocytes." Filed September 21, 1995. U.S. Patent
No. 5,543,318
75. Strobel, ES et al. "Adhesion and migration are differentially regulated
in
hematopoietic progenitor cells by cytokines and extracellular matrix." Blood
(1997) November 1; 90(9):3524-3532.
76. Taylor, D.A. et al. (1998) Nature Med. 4, 929-933.
77. Temple, S. "Opinion: Stem cell plasticity - building the brain of our
dreams."
Nat Rev Neurosci 2001 July;2(7):513-520.
78. Thompson et al. Science 257:868-870 (1992).
79. Tomita, S et al. (1999) Circulation 100(suppl II), II-247-II-256.
80. Vaughn et al. "Incorporating bone marrow transplantation into NCCN
guidelines." (1998) Oncology, 12 (11A): 390-392.
81. Yamaguchi, T.P. et al., "Flk-1, an fit-related receptor tyrosine kinase
is an
early marker for endothelial cell precursors.
Development." (1993) Development 118(2), 489-498.
82. Quaini, F. et al. "Chimerism of the transplanted heart." (2002) N Engl
J
Med.346(1):5-15 N.
83. Anversa, P. and Nadal-Ginard, B., "Myocyte renewal and ventricular
remodelling." Nature. (2002); 415(6868):240-3.
84. Beltrami, A.P. etal., "Chimerism of the transplanted heart." N Engl J
Med.
(2002) 346(1):5-15.
=
85. Reya, T. et al., "Stem cells, cancer, and cancer stem cells." (2001)
Nature
414(6859):105-11.
Jackson, K.A. et al., "Hematopoietic potential of stem cells isolated from
murine skeletal muscle." Proc Natl Acad Sci USA. (1999) 96(25):14482-6.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
86. Orlic, D. et al., "Mobilized bone marrow cells repair the infarcted
heart,
improving function and survival." Proc Natl Acad Sci U S A. (2001)
98(18):10344-9.
87. Blau, H.M. et al., "The evolving concept of a stem cell: entity or
function?"
5 Cell. (2001);105(7):829-41.
88. S. P. Monga, S.P. et al. "Expansion of hepatic and hematopoietic stem
cells
utilizing mouse embryonic liver explants." (2001) Cell Transplant. Jan-Feb;
10(1), 81-89.
89. Weimar, I.S. et al., ",Hepatocyte growth factor/scatter factor (HGF/SF)
is
10 produced by human bone marrow stromal cells and promotes proliferation,
adhesion and survival of human hematopoietic progenitor cells (CD34+)." Exp
HematoL (1998) 26(9):885-94.
90. Yu, C.Z. etal., Stem Cells 16, 66 (1998).
91. Birchmeier, C. and Brohmann, H., Curr. Opin. Cell Biol. 12, 725 (2001).
15 92. Xing, X. etal., Am. J. PathoL 158, 1111 (2001).
93. Hamasuna, R. etal. "Regulation of matrix metalloproteinase-2 (MMP-2)
by
hepatocyte growth factor/scatter factor (HGF/SF) in human glioma cells:
HGF/SF enhances MMP-2 expression and activation accompanying up-
regulation of membrane type-1 MMP." hit J Cancer. (1999) 82(2):274-81.
20 94. Wang, H. and Keiser, J.A., "Hepatocyte growth factor enhances MMP
activity in human endothelial cells." Biochem Biophys Res Commun. 2000
;272(3):900-5.
95. Arsenijevic, Y. et al., "Insulin-like growth factor-I is necessary for
neural
stem cell proliferation and demonstrates distinct actions of epidermal growth
25 factor and fibroblast growth factor-2." J Neurosci. (2001) 21(18):7194-
202
96. Arsenijevic, Y. and Weiss, S., J. Neurosci. "Insulin-like growth factor-
I is a
differentiation factor for postmitotic CNS stem cell-derived neuronal
precursors: distinct actions from those of brain-derived neurotrophic factor."
J
Neurosci. (1998) 18(6):2118-28.
30 97. Brooker, G.J. et al., "Endogenous IGF-1 regulates the neuronal
differentiation
of adult stem cells." J Neurosci Res. (2000) 59(3):332-41.
98. Page, D.L. et al., "Myocardial changes associated with cardiogenic
shock." N
Engl J Med. (1971) 285(3):133-7.
99. Pasumarthi, K.B.S. et al., "Coexpression of mutant p53 and p193 renders
35 embryonic stem cell-derived cardiomyocytes responsive to the growth-
promoting activities of adenoviral ElA." Circ Res. (2001) 88(10):1004-11.
100. Condorelli, G. et aL,"Cardiomyocytes induce endothelial cells to trans-
differentiate into cardiac muscle: implications for myocardium regeneration."
Proc Natl Acad Sci USA. (2001) 98(19):10733-8.
40 101. Beltrami, A.P. etal. "Evidence that human cardiac myocytes divide
after
myocardial infarction." N Engl J Med. (2001) 344(23):1750-7.
102. Jackson, K.A. et al., J. Clin. Invest. (2001) 107, 1395.
103. MacLellan, W.R. and Schneider, M.D. "Genetic dissection of cardiac growth
control pathways." Annu. Rev. PhysioL (2000) 62, 289-319.
45 104. Hidemasa, 0. et al. "Telomerase reverse transcriptase promotes
cardiac
muscle cell proliferation, hypertrophy, and survival." Proc. Natl. Acad. Sci.
USA 98, 10308-10313 (2001).
105. Anversa, P. and Kajstura, J. "Ventricular myocytes are not terminally
differentiated in the adult mammalian heart." Circ. Res. (1998) 83, 1-14.

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
91
106. Rao, M.S. and Mattson, M.P. "Stem cells and aging: expanding the
possibilities. Mech. Ageing Dev. (1998) 122, 713-734.
107. Zaucha, J.M. et al. "Hematopoietic responses to stress conditions in
young
dogs compared with elderly dogs." Blood (2001) 98, 322-327.
108. Gritti, A. et al. "Epidermal and fibroblast growth factors behave as
mitogenic
regulators for a single multipotent stem cell-like population from the
subventricular region of the adult mouse forebrain." J. Neurosci. (1999) 19,
3287-3297.
109. Shihabuddin, L.S. et al., "Adult spinal cord stem cells generate neurons
after
transplantation in the adult dentate gyrus." J. Neurosci. (2000) 20, 8727-
8735.
110. Cheng, W. etal. "Aging does not affect the activation of the myocyte IGF-
1
autocrine system after infarction and ventricular failure in Fischer 344
rats."
Circ. Res. (1996) 78, 536-546.
111. Kajstura, J. et al. "Apoptotic and necrotic myocyte cell deaths are
independent contributing variables of infarct size in rats." Lab. Invest.
(1996)
74, 86-107.
112. Mikawa, T. & Fishman, D.A. "The polyclonal origin of myocyte lineages."
Annu. Rev. Physiol. (1996) 58, 509-521.
113. Stainer, D.Y.R. et al., "Cardiovascular development in zebrafish. I.
Myocardial fate and heart tube formation." Development (1993) 119, 31-40.
114. Hillebrands, J-L. et al. "Origin of neointimal endothelium and a-actin-
positive smooth muscle cells in transplant arteriosclerosis." J. Clin. Invest.
(2001) 107, 1411-1422.
115. Eisenberg, C.A & Bader, D. "QCE-6: a clonal cell line with cardiac
myogenic and endothelial cell potentials." Dev. Biol. (1995) 167, 469-481.
116. Kehat, I. et al. "Human embryonic stem cells can differentiate into
myocytes
with structural and functional properties of myocytes." J. Clin. Invest.
(2001)
108, 407-414.
117. Anderson, D.J. "Stem cells and pattern formation in the nervous system:
the
possible versus the actual." Neuron (2001) 30, 19-35.
118. Lee, J.Y. etal. "Clonal isolation of muscle-derived cells capable of
enhancing
muscle regeneration and bone healing." J. Cell Biol. (2000) 150, 1085-1099.
119. Seale, P. et al. "Pax7 is required for the specification of myogenic
satellite
cells." Cell (2000) 102, 777-786.
120. Broudy, V.C. "Stem cell factor and hematopoiesis." Blood (1997) 90, 1345-
1364.
121. Tropepe, V. et al. "Distinct neural stem cells proliferate in response to
EGF
and FGF developing mouse telencephalon." Dev. Biol. (1999) 208, 166-188.
122. Li, P. et. al. "Myocyte performance during evolution of myocardial
infarction
in rats: effects of propionyl-L-carnitine." Am. J. PhysioL (1995) 208, H1702-
H1713.
123. Beltrami, A.P. et al., Submitted (2002).
124. Bunting, K.D. et al., Blood 96, 902 (2000).
125. Block, G.D. et al., J. Cell Biol. 132, 1133 (1996).
126. Rappolee, D.A. et al., Circ. Res. 78, 1028 (1996).
127. Powell, E.M. et al., Neuron. 30, 79 (2001).
128. Len, A. et al., Circ. Res. 84, 752 (1999).
129. Capasso, J.M. and Anversa, P., Am. J. PhysioL 263, H841 (1992).

CA 02488346 2004-12-03
WO 03/103611
PCT/US03/07731
92
130. Terada, N. et al. Nature, Advanced online publication DOT: nature730,
(2002).
131. Ying, Q-L. et al., Nature, Advanced online publication DOT: nature729,
(2002).

Dessin représentatif
Une figure unique qui représente un dessin illustrant l'invention.
États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Le délai pour l'annulation est expiré 2020-03-12
Représentant commun nommé 2019-10-30
Représentant commun nommé 2019-10-30
Lettre envoyée 2019-03-12
Inactive : TME en retard traitée 2018-09-28
Requête pour le changement d'adresse ou de mode de correspondance reçue 2018-03-28
Lettre envoyée 2018-03-12
Inactive : TME en retard traitée 2017-03-15
Lettre envoyée 2017-03-13
Inactive : TME en retard traitée 2016-03-24
Lettre envoyée 2016-03-14
Inactive : TME en retard traitée 2016-03-11
Lettre envoyée 2015-03-12
Accordé par délivrance 2014-03-11
Inactive : Page couverture publiée 2014-03-10
Préoctroi 2013-12-13
Inactive : Taxe finale reçue 2013-12-13
Un avis d'acceptation est envoyé 2013-06-17
Lettre envoyée 2013-06-17
Un avis d'acceptation est envoyé 2013-06-17
Inactive : Pages reçues à l'acceptation 2013-05-29
Inactive : Lettre officielle - Soutien à l'examen 2013-04-02
Inactive : Approuvée aux fins d'acceptation (AFA) 2013-03-26
Requête visant le maintien en état reçue 2013-02-28
Modification reçue - modification volontaire 2013-02-06
Inactive : Dem. de l'examinateur par.30(2) Règles 2012-08-07
Modification reçue - modification volontaire 2012-05-25
Inactive : Dem. de l'examinateur par.30(2) Règles 2011-11-28
Modification reçue - modification volontaire 2011-06-02
Inactive : Dem. de l'examinateur par.30(2) Règles 2010-12-02
Lettre envoyée 2008-05-12
Modification reçue - modification volontaire 2008-03-12
Exigences pour une requête d'examen - jugée conforme 2008-03-12
Toutes les exigences pour l'examen - jugée conforme 2008-03-12
Requête d'examen reçue 2008-03-12
Inactive : CIB en 1re position 2005-11-22
Inactive : CIB attribuée 2005-11-22
Inactive : CIB enlevée 2005-11-22
Inactive : CIB enlevée 2005-11-22
Inactive : CIB enlevée 2005-11-22
Inactive : CIB enlevée 2005-11-22
Inactive : CIB attribuée 2005-11-22
Inactive : CIB attribuée 2005-11-22
Lettre envoyée 2005-09-06
Inactive : Transfert individuel 2005-07-11
Inactive : Lettre de courtoisie - Preuve 2005-02-22
Inactive : Page couverture publiée 2005-02-21
Inactive : Notice - Entrée phase nat. - Pas de RE 2005-02-17
Inactive : IPRP reçu 2005-02-08
Demande reçue - PCT 2005-01-14
Exigences pour l'entrée dans la phase nationale - jugée conforme 2004-12-03
Demande publiée (accessible au public) 2003-12-18

Historique d'abandonnement

Il n'y a pas d'historique d'abandonnement

Taxes périodiques

Le dernier paiement a été reçu le 2013-02-28

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
NEW YORK MEDICAL COLLEGE
Titulaires antérieures au dossier
PIERO ANVERSA
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document. Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(aaaa-mm-jj) 
Nombre de pages   Taille de l'image (Ko) 
Description 2004-12-02 92 5 476
Dessins 2004-12-02 96 2 670
Abrégé 2004-12-02 2 94
Revendications 2004-12-02 9 288
Dessin représentatif 2004-12-02 1 36
Description 2011-06-01 93 5 553
Revendications 2011-06-01 7 238
Description 2012-05-24 93 5 554
Revendications 2012-05-24 7 218
Revendications 2013-02-05 6 221
Description 2013-05-28 93 5 557
Dessins 2012-05-28 96 2 739
Dessin représentatif 2014-02-03 1 40
Rappel de taxe de maintien due 2005-02-16 1 111
Avis d'entree dans la phase nationale 2005-02-16 1 194
Courtoisie - Certificat d'enregistrement (document(s) connexe(s)) 2005-09-05 1 104
Rappel - requête d'examen 2007-11-13 1 119
Accusé de réception de la requête d'examen 2008-05-11 1 190
Avis du commissaire - Demande jugée acceptable 2013-06-16 1 164
Avis concernant la taxe de maintien 2015-04-22 1 170
Avis concernant la taxe de maintien 2016-03-23 1 170
Quittance d'un paiement en retard 2016-03-23 1 163
Quittance d'un paiement en retard 2016-03-22 1 163
Quittance d'un paiement en retard 2016-03-22 1 162
Quittance d'un paiement en retard 2016-03-23 1 163
Quittance d'un paiement en retard 2018-09-27 1 165
Quittance d'un paiement en retard 2018-09-27 1 165
Avis concernant la taxe de maintien 2017-03-14 1 182
Quittance d'un paiement en retard 2017-03-14 1 164
Quittance d'un paiement en retard 2017-03-14 1 164
Avis concernant la taxe de maintien 2018-04-22 1 178
Avis concernant la taxe de maintien 2019-04-22 1 180
PCT 2004-12-02 3 89
PCT 2004-12-02 5 228
Correspondance 2005-02-16 1 28
Taxes 2008-03-11 1 35
Taxes 2009-02-17 1 36
Taxes 2011-02-17 1 39
Taxes 2012-01-08 1 68
Taxes 2013-02-27 1 68
Correspondance 2013-04-01 1 23
Correspondance 2013-05-28 6 199
Correspondance 2013-12-12 2 78
Taxes 2016-03-23 1 27